You are on page 1of 924

Jane E.

Sykes, BVSc, PhD, DACVIM


Professor
Department of Medicine and Epidemiology
University of California
Davis, California
3251 Riverport Lane
St. Louis, Missouri 63043

CANINE AND FELINE INFECTIOUS DISEASES ISBN: 978-1-4377-0795-3


Copyright © 2014 by Saunders, an imprint of Elsevier Inc.
Figure 58-1 @ owned by UGA Research Foundation.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the publisher. Details on how to seek permission, further information about the
Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance
Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).

Notices
Knowledge and best practice in this field are constantly changing. As new research and experience
broaden our understanding, changes in research methods, professional practices, or medical treat-
ment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evalu-
ating and using any information, methods, compounds, or experiments described herein. In using
such information or methods they should be mindful of their own safety and the safety of others,
including parties for whom they have a professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check the
most current information provided (i) on procedures featured or (ii) by the manufacturer of each
product to be administered to verify the recommended dose or formula, the method and duration
of administration, and contraindications. It is the responsibility of practitioners, relying on their own
experience and knowledge of their patients, to make diagnoses, to determine dosages and the best
treatment for each individual patient, and to take all appropriate safety precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors,
assume any liability for any injury and/or damage to persons or property as a matter of products
liability, negligence or otherwise, or from any use or operation of any methods, products, instruc-
tions, or ideas contained in the material herein.

Library of Congress Cataloging-in-Publication Data


Sykes, Jane E.
Canine and feline infectious diseases / Jane E. Sykes.
p. ; cm.
Includes bibliographical references and index.
ISBN 978-1-4377-0795-3 (hard back)
I. Title.
[DNLM: 1. Communicable Diseases--veterinary. 2. Cat Diseases. 3. Dog Diseases. SF 781]
SF781
636.089’69--dc23 2013010391

Vice President and Publisher: Linda Duncan


Content Strategy Director: Penny Rudolph
Content Development Specialist: Brandi Graham
Publishing Services Manager: Catherine Jackson
Senior Project Manager: Mary Pohlman
Designer: Paula Catalano

Printed in China

Last digit is the print number:  9  8  7  6  5  4  3  2  1


To Terry Nagle, who did countless loads of laundry and cooked
children’s meals while I worked on this book. And to my children,
Henry, Bridget, and Gillian, who through looking over my shoulder
have learned a little about infectious diseases.

Perhaps readers might enjoy seeing some


of what they have learned:

Cryptococcus Antibodies, Phagocytes and Microbes

Dermatophytes Flagellae Virus

Virus Poxvirus Amoeba


Preface

The goal of this first edition of Canine and Feline Infectious well as clinical syndromes (Section 5). A summary box at the
Diseases was to provide a text for veterinary students and cli- beginning of the chapter provides an overview of the disease or
nicians with a strong clinical emphasis. Authorship has been diseases covered. Each chapter contains information on etiology
limited to veterinarians who have had direct and considerable and epidemiology, clinical signs and their pathogenesis; physical
clinical experience with diagnosis and treatment of the diseases examination abnormalities, diagnosis, treatment and prognosis,
described. The chapters have been presented in a consistent and immunity and vaccination, prevention, and public health impli-
logical format with extensive use of tables, photographs, and cations. Some information on the equivalent disease in humans
line drawings, so that veterinarians and students can quickly is provided whenever pertinent. Where relevant, maps and life
find answers to questions they have that relate to infectious dis- cycle drawings are included, and chapters are illustrated with
eases in dogs and cats. photographs of case material. All chapters contain a table that
summarizes the performance, advantages and disadvantages of
Organization the major diagnostic assays available for each disease, and many
chapters also contain a table that provides antimicrobial drug
This book contains two parts and eight sections. The first treatment recommendations and dosing. Key statements in the
part, Basic Principles in the Diagnosis and Management of text are referenced in case the reader desires additional back-
Small Animal Infection, contains three sections: Laboratory ground information, although provision of an exhaustive list
Diagnosis of Infectious Diseases, Antiinfective Therapy, and of references is not in the scope of this book. A few suggested
Basic Principles of Infection Control. The second part, Major readings are provided.
Infectious Diseases and their Etiologic Agents, systematically One (and occasionally two) case example(s) can be found in
discusses specific infectious diseases of dogs and cats in a way a standard format at the end of almost all of the chapters in the
that is practical and relevant to the veterinary clinician. second half of the book. These cases are designed to empha-
The first section, Laboratory Diagnosis of Canine and size key epidemiological and clinical aspects of the disease, the
Feline Infectious Diseases, is co-authored by a small animal application of diagnostic assays, treatment, and prognosis. The
clinician and a clinical microbiologist. It aims to help practic- cases were seen at the authors’ institutions, and many include
ing veterinarians select the best type of test for detection of the results of full laboratory assessments, radiology, cytol-
an infectious disease, collect the most appropriate specimens ogy, and histopathology reports. Unless otherwise specified,
for testing, optimally transport specimens to the laboratory, all urinalyses and urine cultures were performed on specimens
and most importantly, to interpret test results properly. The collected by cystocentesis, and all thoracic radiographic stud-
second section, Antiinfective Therapy, is co-authored by a ies consisted of left and right lateral and ventrodorsal views.
clinician and a veterinary pharmacologist. The goal of this Abdominal ultrasound examinations consisted of complete
section is to help veterinarians use first principles to select studies of intraabdominal structures. Because these are “real-
the most appropriate drug for treatment of a suspected or world” examples, diagnosis, management, and outcomes are
confirmed infectious disease, understand the mechanisms not always perfect or ideal. Nevertheless, this can provide
of action and adverse effects of various antimicrobial drugs insight into real-life problems that may confound accurate
used in practice, and utilize treatment regimes that minimize diagnosis and management of the disease, such as misdiag-
development of antimicrobial resistance. The third section, nosis when an infectious disease resembles other infectious or
Basic Principles for Infection Control, is co-authored by non-infectious diseases on the differential diagnosis list. Other
veterinary clinicians who have extensive practical experi- problems illustrated include limitations of the sensitivity or
ence with infectious disease control in veterinary hospitals. specificity of diagnostic tests used, or hurdles imposed by the
Practical protocols for the management of canine and feline availability of client financial resources. A comment section at
patients suspected to have infectious diseases are provided, the end of the case provides the author(s’) perspective on the
including handling, disinfection, isolation, and vaccination case. It should be recognized that infectious diseases can vary
protocols. widely in their clinical presentation and response to treatment,
The second half of this book addresses specific infectious so each case just provides one “view” of the infectious disease
diseases of dogs and cats, which are grouped into viral, bacte- described in the chapter.
rial, fungal, and protozoal diseases (Sections 1 through 4) as Jane E. Sykes

vii
Acknowledgments

I am incredibly grateful to the huge number of veterinarians, number of complicated maps and life cycle drawings into crisp,
residents, and veterinary students worldwide who have encour- clean diagrams that are easy to understand and remember. I
aged me to write and edit this book. I would particularly like to have also been fortunate to work with a phenomenal team at
acknowledge all of my colleagues at the University of ­California, Elsevier who were consistently encouraging, efficient, and made
Davis, who over the years provided a large number of the care- every effort to be accommodating. I would like to thank my
fully collected and valuable images in this book. Special men- internal medicine and infectious disease colleagues worldwide,
tions go to Barbara Byrne, Patricia Pesavento, and Eileen Samitz many of whom contributed to chapters for this book, for inspir-
who spent much additional time to prepare or provide material ing me to have a passion for the study of small animal internal
for photography for this book. medicine and infectious diseases. Last but not least I would like
I would like to acknowledge the illustrator for the book, Ted to acknowledge my family, who cheered me on the whole way.
Huff, who has performed the amazing task of converting a large Jane E. Sykes

viii
Contributors

Gad Baneth, DVM, PhD, DECVCP Leah A. Cohn, DVM, PhD, DACVIM
Professor of Veterinary Medicine Professor, Small Animal Internal Medicine
The Robert H. Smith Faculty of Agriculture, Food and Department of Veterinary Medicine and Surgery
Environment College of Veterinary Medicine
Koret School of Veterinary Medicine University of Missouri
The Hebrew University of Jerusalem Columbia, Missouri
Rehovot, Israel Cytauxzoonosis
Leishmaniosis
Sarah D. Cramer, DVM, DACVP
Stephen C. Barr, BVSc, MVS, PhD, DACVIM Cancer Research Training Fellow
Professor of Medicine Comparative Biomedical Scientist Training Program
Chief, Section of Small Animal Medicine National Cancer Institute
College of Veterinary Medicine National Institutes of Health
Cornell University Bethesda, Maryland
Ithaca, New York Pseudorabies
Trypanosomiasis
Autumn P. Davidson, DVM, MS, DACVIM
Malcolm Bennett, BVSc, PhD, MRCVS, FRCPath Clinical Professor
Professor of Veterinary Pathology, Infection, and Global Department of Medicine and Epidemiology
Health School of Veterinary Medicine
Department of Veterinary Pathology University of California
University of Liverpool Davis, California;
Liverpool, United Kingdom Staff Internist
Feline Poxvirus Infections Internal Medicine/Reproduction
PetCare Veterinary Hospital
Adam J. Birkenheuer, DVM, DACVIM, PhD Santa Rosa, California
Associate Professor Canine Herpesvirus Infection
Department of Clinical Sciences Canine Brucellosis
College of Veterinary Medicine
North Carolina University Steven Epstein, DVM, DACVECC
Raleigh, North Carolina Assistant Professor of Clinical Small Animal Emergency and
Babesiosis Critical Care
Department of Veterinary Surgical and Radiological Sciences
Edward B. Breitschwerdt, DVM, DACVIM School of Veterinary Medicine
Professor of Medicine and Infectious Diseases University of California
Department of Clinical Sciences Davis, California
College of Veterinary Medicine Infections of the Cardiovascular System
North Carolina State University
Raleigh, North Carolina Janet E. Foley, DVM, PhD
Rocky Mountain Spotted Fever Professor
Department of Veterinary Medicine and Epidemiology
Bruno B. Chomel, DVM, PhD School of Veterinary Medicine
Professor University of California
Population Health & Reproduction Davis, California
University of California Anaplasmosis
Davis, California
Rabies
Bartonellosis
Yersinia pestis (Plague) and Other Yersinioses
Tularemia

iii
iv CONTRIBUTORS

Craig E. Greene, DVM, MS, DACVIM Remo Lobetti, BVSc, MMedVet (Med), PhD, DECVIM
Professor Emeritus and Josiah Meigs Distinguished Teaching (Internal Medicine)
Professor Internist
Departments of Infectious Diseases and Small Animal Medicine Bryanston Veterinary Hospital
College of Veterinary Medicine Bryanston, South Africa
The University of Georgia Pneumocystosis
Athens, Georgia
Bacterial Meningitis Jennifer A. Luff, VMD, DACVP
Veterinary Medicine
Amy M. Grooters, DVM, DACVIM Department of Pathology, Microbiology, and Immunology
Professor, Companion Animal Medicine School of Veterinary Medicine
Department of Veterinary Clinical Sciences University of California
School of Veterinary Medicine Davis, California
Louisiana State University Viral Papillomatosis
Baton Rouge, Louisiana
Miscellaneous Fungal Diseases Richard Malik, DVSc, DipVetAn, MVetClinStud, PhD,
Pythiosis, Lagenidiosis, and Zygomycosis FACVSc, FASM
Valentine Charlton Veterinary Specialist
Danièlle A. Gunn-Moore, BSc, BVM&S, PhD, MACVSc, Centre for Veterinary Education
MRCVS, RCVS University of Sydney
Professor of Feline Medicine Sydney, Australia
Head of Companion Animal Sciences Cryptococcosis
Royal (Dick) School of Veterinary Studies
The University of Edinburgh Stanley L. Marks, BVSc, PhD, DACVIM (Internal Medicine,
Easter Bush, Midlothian, United Kingdom Oncology), DACVN
Mycobacterial Infections Professor
Department of Medicine and Epidemiology School of
Katrin Hartmann, Dr.med.vet, DMVH, DECVIM-CA Veterinary Medicine
Professor University of California
Center of Clinical Veterinary Sciences Davis, California
Clinic of Small Animal Medicine Salmonellosis
Ludwig-Maximilians-Universität, München Enteric Escherichia coli Infections
Munich, Germany Campylobacteriosis
Feline Leukemia Virus Infection Enteric Clostridial Infections
Gastric Helicobacter-like Infections
Amy S. Kapatkin, DVM, MS
Associate Professor of Orthopedic Surgery Lindsay K. Merkel, DVM, DACVIM
Department of Surgical and Radiological Sciences Internal Medicine Specialist
School of Veterinary Medicine Veterinary Medical Center
University of California College of Veterinary Medicine
Davis, California University of Minnesota
Osteomyelitis, Discospondylitis, and Infectious Arthritis St. Paul, Minnesota
Blastomycosis
Linda Kidd, DVM, PhD, DACVIM
Assistant Professor Terry M. Nagle, BVSc, MACVSc, DACVD
Department of Small Animal Internal Medicine Northern California Veterinary Specialist
College of Veterinary Medicine Emergency & Referral Center
Western University of Health Sciences Sacramento, California
Pomona, California Malassezia Infections
Rocky Mountain Spotted Fever Pyoderma, Otitis Externa, and Otitis Media

Michael R. Lappin, DVM, PhD, DACVIM Jacqueline M. Norris, BVSc, MVS, PhD
Professor of Infectious Disease Associate Professor of Veterinary Microbiology
Department of Clinical Sciences Faculty of Veterinary Science
College of Veterinary Medicine and Biomedical Sciences University of Sydney
Colorado State University Sydney, Australia
Fort Collins, Colorado Coxiellosis and Q Fever
Toxoplasmosis
Giardiasis
Trichomoniasis
Cryptosporidiosis
Isosporiasis
CONTRIBUTORS v

Catherine A. Outerbridge, MVSc, DVM Séverine Tasker, BSc, BVSc, PhD, PGCert (HE), DSAM,
Associate Professor of Dermatology DECVIM-CA, MRCVS
Department of Medicine and Epidemiology Senior Lecturer in Small Animal Medicine
School of Veterinary Medicine School of Veterinary Sciences
University of California University of Bristol
Davis, California Bristol, Avon, United Kingdom
Dermatophytosis Hemoplasma Infections

Mark G. Papich, DVM, MS, DACVCP Nancy Vincent-Johnson, DVM, MS, DACVIM (SAIM),
Professor DACVPM
Department of Molecular Biomedical Sciences Veterinary Medical Officer
College of Veterinary Medicine Chief Clinical Services
North Carolina State University Fort Belvoir Veterinary Treatment Facility
Raleigh, North Carolina United States Army Veterinary Corps
Principles of Antiinfective Therapy Fort Belvoir, Virginia
Antiviral and Immunomodulatory Drugs Canine and Feline Hepatozoonosis
Antibacterial Drugs
Antifungal Drugs J. Scott Weese, DVM, DVSc, DACVIM
Antiprotozoal Drugs Professor
Department of Clinical Studies (Pathobiology)
Christine A. Petersen, DVM, PhD University of Guelph
Associate Professor in Veterinary Pathology Guelph, Ontario
Petersen and Jones Laboratory Infection Control Programs for Dogs and Cats
College of Veterinary Medicine
Iowa State University Jodi L. Westropp, DVM, PhD, DACVIM
Ames, Iowa Associate Professor
Leishmaniosis Department of Medicine and Epidemiology
School of Veterinary Medicine
Shelley C. Rankin, BSc (Hons), PhD University of California
Associate Professor (CE) of Microbiology Davis, California
New Bolton Center Bacterial Infections of the Genitourinary Tract
School of Veterinary Medicine
University of Pennsylvania Stephen D. White, DVM, DACVD
Philadelphia, Pennsylvania Professor
Isolation in Cell Culture Department of Medicine and Epidemiology
Immunoassays School of Veterinary Medicine
Isolation and Identification of Aerobic and Anaerobic Bacteria University of California
Isolation and Identification of Fungi Davis, California
Malassezia Infections
Ashley B. Saunders, DVM, DACVIM (Cardiology) Pyoderma, Otitis Externa, and Otitis Media
Assistant Professor
Department of Small Animal Clinical Sciences
College of Veterinary Medicine and Biomedical Sciences
Texas A&M University
College Station, Texas
Trypanosomiasis

Joseph Taboada, DVM, DACVIM (Internal Medicine)


Professor and Associate Dean
Department of Veterinary Clinical Sciences School of
Veterinary Medicine
Louisiana State University
Baton Rouge, Lousiana
Histoplasmosis
SECTION 1
Laboratory Diagnosis of Canine
and Feline Infectious Diseases
Jane E. Sykes  and  Shelley C. Rankin

CHAPTER 1

Isolation in Cell Culture


Jane E. Sykes and Shelley C. Rankin

w KEY POINTS
• W  ith the increasing availability of nucleic acid–based testing, • S pecimens are inoculated onto monolayers, and the infecting
cell culture is decreasingly used for diagnosis of infections organism is identified based on the presence of characteristic
caused by obligate intracellular pathogens in dogs and cats. cytopathic effect after a predictable incubation period, with or
• Cell culture remains an important technique for (a) confirma- without confirmatory antigen staining, electron microscopy, or
tion of a diagnosis when the results of molecular testing or nucleic acid testing.
serology are unavailable or equivocal; (b) pathogen discovery; • False-negative results may occur as a result of inadequate spec-
and (c) vaccine manufacture. For some pathogens, cell cul- imen collection, deterioration of organisms during transport, or
ture is the most sensitive and specific method for organism culture contamination with bacteria or fungi.
detection. • Positive results do not imply that the organism detected is the
• Before collection of specimens, veterinary clinicians should cause of an animal’s signs, because some organisms can be
communicate with the laboratory that is to perform the culture present without causing disease. This is especially the case for
to discuss the patient signalment, history, immune status, travel animals with respiratory or gastrointestinal disease.
history, nature of the suspected infection, and number of ani-
mals affected.

INTRODUCTION distemper, canine adenovirus infections, parvovirus infections,


rabies, and feline viral and chlamydial respiratory tract disease.
Cell culture refers to the culture of nucleated (eukaryotic) cells Veterinary clinicians should remain aware of situations where cell
under controlled conditions within the laboratory. Infectious culture may be the best technique to identify the presence of an
agents that require living host cells for replication can only be infectious agent and the optimum methods for collection and
isolated in cell culture. With the advent of molecular diagnostic submission of specimens. Knowledge of cell culture methods can
assays based on nucleic acid detection, cell culture is being used help veterinary clinicians to submit the optimum specimens and
less often for routine clinical diagnostic purposes, because of the to understand laboratory turnaround times, potential complica-
long turnaround times (days to weeks), cost, and requirement for tions, and how to interpret results.
significant technical expertise to perform cell culture and interpret
results (Table 1-1). Nevertheless, isolation of viral and intracellu- Specimen Collection and Transport
lar bacterial and protozoal pathogens in cell culture remains an
important technique for the discovery of new pathogens, iden- Although cell culture can be used to propagate intracellular
tification of organisms involved in disease when the results of bacteria and protozoa, it is most often used by clinicians for
molecular testing or serology are unavailable or equivocal, the the diagnosis of viral infections. Active communication between
propagation of isolates for research purposes, the generation of the clinician and the laboratory that performs viral isolation is
organisms for vaccination purposes, and the establishment of recommended. Successful detection of viruses is highly depen-
the efficacy of novel antimicrobial drugs. Vaccines for dogs and dent on (a) collecting the appropriate specimens, (b) the tim-
cats that are propagated in cell culture include those for canine ing of specimen collection, and (c) rapid and proper specimen

2
CHAPTER 1  Isolation in Cell Culture 3

TABLE 1-1 BOX 1-1


Alternatives to Cell Culture for Diagnosis of Obligate Factors That Should Be Discussed with the Laboratory
Intracellular Pathogens That Infect Dogs and Cats before Collection of Specimens for Pathogen Isolation in
Cell Culture
System Affected and Most Other Diagnostic Tests
Common Agents Available
Patient species, breed, age, and environment
Respiratory Tract Number of animals affected
Canine respiratory coronavirus RT-PCR History and clinical signs
Canine adenovirus-2 PCR Immune status of the patient
Influenza viruses RT-PCR, antibody Geographic location and travel history
Canine parainfluenza virus detection Suspected infectious agents
Canine distemper virus RT-PCR Timing of specimen collection
Canine herpesvirus RT-PCR, antigen detection Type and amount of specimen to be collected
Feline herpesvirus-1 using IFA Transport conditions, including timing and method of
Feline calicivirus PCR transportation
PCR, IHC
RT-PCR, IHC

Eye coronavirus (FCoV), are difficult to isolate in cell culture or


Chlamydia felis PCR grow slowly, whereas others, such as feline calicivirus (FCV),
replicate readily and rapidly in cell culture, and the sensitivity of
Central Nervous System cell culture is high. Viruses differ in respect to the cell type they
Canine distemper virus RT-PCR, direct IFA, CSF prefer to replicate within. As a result, specimens should be sent
West Nile virus antibody detection to the laboratory with information on the specific viruses that
Encephalitis viruses RT-PCR are suspected.
RT-PCR The timing of specimen collection is particularly important
for viral infections. Specimens should be collected as early as
Gastrointestinal Tract possible following the onset of clinical signs, optimally within
Coronaviruses PCR the first week, because viral shedding may commence before
Canine distemper virus PCR, direct IFA/IHC the onset of signs and continue for only a few days. The dura-
Canine and feline parvovirus PCR, IHC tion of viral shedding depends on the type of virus and the
anatomic site sampled. When multiple animals are affected,
Genital collection of specimens from more than one animal may
Canine herpesvirus PCR increase the chance that an isolate will be obtained. If possible,
antibody testing using acute and convalescent phase serology
Congenital and Perinatal should be performed concurrently to help confirm the diagno-
Canine herpesvirus PCR sis (see Chapter 2).
Feline herpesvirus-1 PCR Selection of the best specimen and collection site for culture
is optimized based on knowledge of the pathogenesis of the
Blood infectious agent involved, because the optimum specimen col-
Feline leukemia virus PCR, antigen detection lection site may not be the site where clinical signs are most
Feline immunodeficiency virus PCR, antibody detection severe. Attempts should be made during specimen collection
Anaplasma phagocytophilum PCR, antibody detection to prevent contamination of the specimen with normal flora,
Rickettsia rickettsii PCR, direct IFA on skin although this is not always possible. Specimen size should also
Ehrlichia canis biopsies, antibody detec- be maximized (for example, at least 5 mL of blood, body flu-
tion ids, or lavage specimens, and ideally 8 to 10 mL of blood) to
PCR, antibody detection increase the chance of a positive isolation. In general, nasal or
nasopharyngeal washes have been preferred over nasal swabs
CSF, Cerebrospinal fluid; IFA, fluorescent antibody; IHC, immunohis- in human patients for isolation of respiratory viruses, but one
tochemistry; PCR, polymerase chain reaction; RT, reverse transcriptase.
study showed that nasal swab specimens were just as sensitive
as nasopharyngeal washes for isolation of most respiratory
transport and processing. Thus the actions of the veterinary cli- viruses.1 Nasal or oropharyngeal swab specimens are collected
nician play a critical role in ensuring positive test results when by placing a long-shafted swab in the area to be sampled, rotat-
a virus is present. ing the swab against the mucosa, and allowing the secretions to
The clinician should discuss with the laboratory what types be absorbed for approximately 5 to 10 seconds.
of viruses are suspected in light of the animal’s clinical presen- Swabs and small tissue specimens for virus isolation should
tation. The patient signalment, history, clinical signs, immune be placed in buffered virus transport medium, which contains
status, travel history, and number of animals affected should antibiotics and protein. This can be obtained from the labora-
be discussed to generate conclusions regarding the nature of tory or purchased from other commercial sources. It is impor-
the suspected infection (Box 1-1). Some viruses, such as feline tant that the medium used has not reached its expiry date.
4 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

TABLE 1-2
Specimen Collection Guide for Diagnosis of Viral and Intracellular Bacterial Infections of Companion Animals
System Affected Possible Agents Specimen Type
Respiratory Dogs: coronaviruses, canine adenovirus, Oropharyngeal swabs
tract influenza viruses, parainfluenza virus, Nasal flushes, transtracheal wash or bronchoalveolar lavage speci-
CDV, canine herpesvirus mens: ideally 5 to 10 mL of fluid
Cats: FHV-1, FCV, influenza viruses, Lung tissue obtained at biopsy or necropsy, including an area adja-
FCoV cent to affected tissue
Eye Dogs: canine herpesvirus, canine Conjunctival swab, scraping or biopsy
adenovirus
Cats: FHV-1, FCV, Chlamydia felis
Central nervous Dogs: CDV, West Nile virus, Cerebrospinal fluid: ideally at least 0.5 to 1 mL
system arboviruses Blood: 8 to 10 mL
Brain at necropsy
Gastrointestinal Dogs: CDV, CPV, rotaviruses, canine Feces: ideally an olive-sized portion of formed feces or 10 mL of
tract coronavirus liquid stool
Cats: FCoV, FCV, FeLV, rotaviruses, Intestinal biopsies obtained using endoscopy or surgery, or intesti-
toroviruses nal tissue obtained at necropsy
Genital Dogs: canine herpesvirus Vesicle scrapings, vaginal swabs
Cats: Chlamydia felis
Congenital and Dogs: canine herpesvirus Blood, tissues obtained at necropsy
perinatal Cats: FHV-1, FeLV
Blood Dogs: Anaplasma phagocytophilum, Blood: ideally 8 to 10 mL
Rickettsia rickettsii, Ehrlichia canis
Cats: FeLV, FIV, FCoV

CDV, Canine distemper virus; CPV, canine parvovirus; FCoV, feline coronavirus; FCV, feline calicivirus; FeLV, feline leukemia virus; FHV-1, feline
herpesvirus-1; FIV, feline immunodeficiency virus.

Liquid specimens such as blood, cerebrospinal fluid, and Category B. Category A infectious substances are those capa-
bronchoalveolar lavage fluid do not need to be placed in trans- ble of causing permanent disability or life-threatening or fatal
port media. Blood samples should be collected using sterile tech- disease in otherwise healthy animals and humans.2 Most speci-
nique, with antiseptic preparation of the site of venipuncture, mens submitted by veterinarians fall under Category B, which
and can be submitted in EDTA anticoagulant tubes. are those that do not fall under the criteria for inclusion in Cat-
All specimens should be refrigerated on collection and trans- egory A. Updated documents providing guidance on regulations
ported as quickly as possible (preferably within 24 hours) to the for the transport of infectious substances are provided online by
laboratory, because delayed transport can lead to loss of organ- the World Health Organization (WHO).2 Import permits may be
ism viability. If delays in excess of 2 to 3 days are anticipated, required for interstate and international transportation.
the specimen can be frozen. Freezing should be avoided when-
ever possible, as it may lead to dramatic loss of virus viability. Diagnostic Methods
If freezing is unavoidable, freezing at −70° C is preferable to
freezing at −20° C, and shipping on dry ice is preferable, if pos- Maintenance of Cell Cultures in the Laboratory
sible. The laboratory’s submission guide should be checked for In general, cells are grown as a monolayer on a plastic plate. The
specimen handling recommendations. cells in the monolayer can be derived directly from an animal
Table 1-2 provides a guide to the recommended specimen (primary cell culture), which tend to have a limited life span, or
types for isolation of viruses or obligate intracellular bacteria they may be immortalized (continuous cell lines). Primary cell
from companion animals. Specimens should be labeled with the cultures are needed for the isolation of some viruses, because
patient data, the site(s) from which the specimen(s) was collected, the cells more closely resemble those present in  vivo, and the
specific organisms suspected, and the time and date of specimen replication of these viruses occurs more efficiently in primary
collection. Contained specimens should be placed inside leak- cell lines than in continuous cell lines. Further subculture of pri-
proof triple packaging and transported on wet ice or cold packs mary cell lines often reduces their sensitivity to viral infection.
to the laboratory, especially if transport is expected to take lon- Primary cell cultures are generated by placing tissues in cell cul-
ger than 1 hour. Absorbent materials should be placed within the ture media, often after treatment of the tissue with an enzyme
secondary container in order to absorb any spills. If specimens are such as trypsin or collagenase. Primary white blood cell cultures
to be shipped, the specimen must be labeled and handled accord- (such as peripheral blood mononuclear cell cultures) are gen-
ing to governmental and International Air Transport Associa- erated by separation of the white cells from the other cellular
tion (IATA) regulations for shipping materials known to contain elements using density gradient centrifugation, and adding them
infectious substances, which are categorized as Category A or to a culture medium. Ficoll, a highly branched polysaccharide,
CHAPTER 1  Isolation in Cell Culture 5

TABLE 1-3
Examples of Continuous Cell Lines Used for Isolation of Viruses
and Intracellular Bacteria That Infect Dogs and Cats
Cell Line Cell Origin Pathogen(s)
Vero cells; African Green CDV11,12
recombinant monkey renal Rickettsia rickettsii13
Vero-SLAM epithelial cells Toxoplasma gondii14
cells
Madin-Darby Kidney CDV8,15
canine kidney Canine adenovirus8,15
cells (MDCK) Canine herpesvirus-18,15
Parvoviruses8,16
Canine parainfluenza A
virus8
Canine calicivirus4
Rotaviruses17
Influenza viruses18
FeLV19
Neospora caninum20
Crandell-Reese Fetal kidney FHV-121
feline kidney FCV21,22
cells FCoV23
Parvoviruses24
FIV25
HL-60 Human leukemia Anaplasma phagocyto-
philum26
A-72 Canine fibroma Canine adenovirus27
Canine coronavirus27
Canine parainfluenza B
virus27 FIGURE 1-1  A, Plastic flask that contains cell culture medium. B, A confluent cell
Canine herpesvirus27 monolayer is present on the bottom of the flask and can be visualized through the top of
McCoy Mouse fibroblast Chlamydia felis28 the flask using a binocular inverted microscope.
FCWF Felis catus whole FCoV29
different viruses prefer to replicate in differing cell types. Mixed
fetus, has FHV-130
cell cultures are also now available commercially to simultane-
­characteristics
ously facilitate isolation of multiple different viral pathogens.
of macrophages
Cells for cell culture are stored in the laboratory in liquid
DH-82 Monocyte/macro- Ehrlichia canis31 nitrogen tanks. The cells are thawed, dispersed in cell culture
phage medium, and allowed to settle on the bottom of a plastic flask
(Figure 1-1). The cell culture medium keeps the cells moist and
CDV, Canine distemper virus; FCoV, feline coronavirus; FCV, feline
calicivirus; FeLV, feline leukemia virus; FHV-1, feline herpesvirus-1; provides the cells with nutrients. Minimum essential medium
FIV, feline immunodeficiency virus. (MEM), also known as Eagle’s minimum essential medium, and
Dulbecco’s medium are examples of widely used synthetic cell
culture media. The cell culture medium contains a balanced salt
is an example of a medium used commonly for density gradient solution, essential amino acids, glucose, vitamins, and a bicar-
centrifugation. Primary cell cultures have been used widely for bonate buffering system. Variations of MEM are available, some
the isolation of intracellular pathogens of dogs and cats.3-6 of which contain nonessential amino acids, the pH indicator phe-
Low-passage cell lines remain viable and sensitive to viral nol red, and the pH buffering agent HEPES (4-(2-hydroxyethyl)-
infections for 20 to 50 passages. Continuous cell lines are the type 1-piperazineethanesulfonic acid), which helps to maintain a
of cell line used most commonly for diagnostic, research, and physiologic pH despite an increasing concentration of CO2 that
commercial purposes. These are derived from cancer cells (such as is produced as a result of cellular respiration. The medium often
the widely used HeLa cell line, derived from human cervical can- requires supplementation with animal serum, most commonly
cer cells of a patient named Henrietta Lacks),7 or they result from fetal calf serum, which helps the cells to attach to and spread
experimental induction of cellular mutations (for example, using on the plate, although serum-free medium has also been used
a carcinogen). Continuous cell lines representing a wide variety successfully for culture of canine and feline viruses.8 Media for
of cell types are available from commercial suppliers (Table 1-3). cell culture growth differ from media for maintenance of cells in
Laboratories that perform virus isolation for disease diagnosis culture, the former generally containing a higher concentration
may need to simultaneously inoculate multiple cell lines, because of animal serum (10% versus 2% for maintenance medium).
6 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

To prevent bacterial or fungal contamination when viral Inoculation of Cell Cultures


pathogens are cultured, careful attention to sterile technique is Infection of a cell monolayer is accomplished by removal of
required. Manipulation of cells in the flask is performed in a overlying medium and inoculation of the monolayer with a sus-
laminar flow hood, and the culture medium is sometimes sup- pension of viral or bacterial organisms. If tissue specimens are
plemented with antibiotics (generally ampicillin or an amino- provided for culture, the specimens are homogenized in culture
glycoside), with or without an antifungal agent (amphotericin B medium before they are inoculated onto the monolayer. Speci-
or nystatin). Cell culture flasks are placed in a humidified incu- mens may first be centrifuged to remove cell debris and bacte-
bator that maintains temperature usually at 37°C, and a gas ria. Passage through a filter can also be used before inoculation
mixture that typically contains 5% CO2. to remove bacteria. The inoculum is added to the monolayer.
The cell culture flasks are removed from the incubator and Organisms within the inoculum are then allowed to settle on
examined daily using an inverted binocular tissue culture micro- the monolayer for approximately 1 hour before the residual
scope (see Figure 1-1), which allows examination of unstained inoculum is removed and fresh maintenance medium is added.
cells through the bottom of the flask. The medium is replaced For isolation of virus, the monolayer is then examined daily
with fresh medium if the pH indicator suggests it is becoming for evidence of cytopathic effect (CPE). Medium is replaced
too acidic. Once the cells have multiplied sufficiently to form a weekly or biweekly. CPE refers to the cellular changes induced
semiconfluent monolayer, the cells can either be passaged or be by viral replication in the cell culture monolayer and includes
allowed to reach confluence before inoculation with specimen. cell lysis or cell fusion (syncytium formation). The presence of a
In order to passage the cells, the medium is removed, and a small particular viral pathogen is indicated by the appearance of char-
volume of trypsin or EDTA solution is added to the monolayer. acteristic CPE for that pathogen after a predicted incubation
The monolayer is incubated with the solution for several min- period (Figure 1-2). For example, CPE induced by FCV is char-
utes, after which the cells detach from the flask and can be resus- acterized by cell rounding and detachment that can occur within
pended in medium, which is then added to new flasks. 24 hours, and within 16 hours for highly virulent strains.10
Occasionally, cell lines become cross-contaminated with other The presence of CPE is confirmed through the comparison of
cell lines. The HeLa cell line is an example of a prolific and hardy inoculated cultures to uninoculated (control) cultures. Exami-
cell line that commonly contaminates other cultures. Major cell nation of the cell monolayer using light microscopy after fixa-
line repositories such as the American Type Culture Collection tion and staining can reveal additional diagnostic features, such
(ATCC) use genetic typing methods to verify the identity of the as the presence of inclusion bodies and syncytium formation.
cells in their collection. Cell lines can also become easily con- Some viruses are relatively noncytopathogenic. In some cases,
taminated with Mycoplasma spp., which pass through filters the presence of these viruses can be demonstrated by adding
used to exclude other bacteria. The presence of contaminating washed erythrocytes to the monolayer and examination of
Mycoplasma spp. can interfere with replication of other patho- the monolayer for evidence of hemadsorption (see Figure 1-2,
gens and cause alterations of cellular morphology. Mycoplasma B). Hemadsorption results from incorporation of viral surface
contamination is detected using stains such as Hoechst stain, hemagglutinin molecules into the plasma membranes of the cell
culture for Mycoplasma, and commercially available PCR assays culture monolayer. Examples of hemadsorbing viruses that pro-
that specifically detect Mycoplasma DNA for the purpose of cell duce minimal CPE include influenza and parainfluenza viruses.
culture quality assurance.9 Other, more sensitive and specific methods for identification of

A B
FIGURE 1-2  A, Cytopathic effects produced by a herpesvirus as viewed in the laboratory. Note the focal areas of rounded and detached cells. B, Hemadsorption: erythrocytes adsorb
to infected cells that have incorporated hemagglutinin into the plasma membrane. Magnification ×60. (Courtesy of Jack I; from MacLachlan NJ, Dubovi EJ eds. Fenner’s veterinary virology,
4 ed. New York: Academic Press, 2011.)
CHAPTER 1  Isolation in Cell Culture 7

A B C

D E F
FIGURE 1-5  Determination of the concentration of infectious virus using a plaque
assay. Vero cell monolayers were inoculated with serial 10-fold dilutions of vesicular sto-
FIGURE 1-3  Indirect fluorescent antibody detection of noncytopathic virus infected matitis virus. After a 1-hour adsorption period, cultures were overlayed with 0.75% aga-
cells. A cell monolayer exposed to virus for 72 hours was fixed with cold acetone. Fixed rose in cell culture medium containing 5% fetal bovine serum. Cultures were incubated for
cells were stained with a mouse monoclonal antibody specific for bovine viral diarrhea 3 days at 37°C in a 5% humidified CO2 atmosphere. The agarose overlay was removed and
virus (BVDV) followed by a goat anti-mouse serum tagged with fluorescein isothiocyanate. cultures fixed and stained with 0.75% crystal violet in 10% buffered formalin. A, Control
(From Maclachlan NJ, Dubovi EJ. Fenner’s Veterinary Virology, 4 ed. New York: Academic culture. B-F, serial 10-fold dilution of virus: B, 10−3; C, 10−4; D, 10−5; E, 10−6; F, 10−7.
Press, 2011.) (From Maclachlan NJ, Dubovi EJ. Fenner’s Veterinary Virology, 4 ed. New York: Academic
Press, 2011.)

antibodies, removed from the vials, and examined with a fluo-


rescence microscope.

Plaque Assays
Overlay of the monolayer with agar or methylcellulose after
inoculation with virus minimizes subsequent viral movement
through the monolayer and restricts damage by each replicating
viral particle to a small area. This results in the formation of
“holes” in the monolayer, or plaques (Figure 1-5). The mono-
layer can then be stained and the number of plaques can be
counted in order to obtain information regarding the amount
of virus in the inoculum. Plaque assays are commonly used by
researchers to assess the efficacy of antiviral treatments, through
reduction of plaque formation.

FIGURE 1-4  Schematic of the shell vial virus isolation technique. (Modified from Laboratory Safety Concerns That Relate
Shelhamer JH, Gill VJ, Quinn TC, et al. The laboratory evaluation of opportunistic pulmo- to Cell Culture
nary infections. Ann Intern Med 1996;124(6):585-599.) The processing of specimens in a biological safety cabinet not
only serves to protect cultures from contamination but also
acts to protect the laboratory worker from ­laboratory-acquired
an organism that has infected a cell culture monolayer include infections. Most viruses grown in veterinary diagnostic labora-
fixation and staining of the monolayer with fluorescent anti- tories are classified as biosafety level 2 (BSL 2) agents. Biosafety
bodies that are specific for certain viruses (Figure 1-3), use of levels range from 1 through 4 (Table 1-4). In the United States,
nucleic acid detection techniques such as PCR, or use of electron the Centers for Disease Control and Prevention (CDC) assigns
microscopy. these levels. It is important that veterinary clinicians be aware
For spin-amplified cell culture, the monolayer is centrifuged that isolation of hazardous pathogens can only be conducted in
at low speed after inoculation. This can enhance recovery of specially designed and accredited laboratories.
certain viruses and intracellular bacterial pathogens such as
chlamydiae. Either an entire plate of cell culture wells can be Interpretation of Results
spun in a plate centrifuge, or shell vials can be used (also known
as the shell vial technique). Shell vials are small, flat-bottomed Negative Results
bottles (Figure 1-4). A monolayer is grown on a glass coverslip Reasons for negative test results using virus isolation are numer-
at the bottom of the vial, and after inoculation, the vials are ous, so the clinician should not assume that a virus is not pres-
centrifuged at low speed. Using this method, fluorescent anti- ent when a negative test result is obtained (Box 1-2). Negative
body staining for viral or chlamydial antigen is used to detect test results can occur as a result of inadequate specimen size,
the pathogen before CPE occurs. After 48 to 72 hours, the cov- lack of viral shedding by the animal at the time of specimen col-
erslips are fixed, stained with virus-specific fluorescent-labeled lection, the binding of antibodies to viruses within the specimen,
8 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

TABLE 1-4
Biosafety Levels Set for Diagnosis of Laboratory Practices
Level Risk Examples Example Precautions
BSL 1 Minimal potential hazard to human Canine adenovirus-1 Gloves, facial protection. Standard microbiologic
health and the environment Nonpathogenic practices using bench-top techniques. Routine
Escherichia coli decontamination practices (hand washing, routine
bench disinfection, autoclaving of infectious waste).
BSL 2 Moderate potential hazard to Most veterinary Access to the laboratory restricted when work is taking
human health and the environment. viruses, including place; extreme precautions with sharp contaminated
Organisms cause mild disease or are influenza viruses materials; use of appropriate biosafety cabinets
difficult to contract as laboratory when generation of aerosols possible. No require-
aerosols. ment for directional airflow into the laboratory.
BSL 3 Dangerous agents that can be West Nile virus Laboratory is located away from high-traffic areas.
transmitted by aerosol within the Equine encephalitis Restricted laboratory access when work in progress;
laboratory but for which effective viruses double door entry, ventilation providing airflow
vaccines or treatments exist. Rickettsia rickettsii into the room, exhaust air not recirculated; special
Coxiella burnetii practices and protective clothing for BSL 3, including
Mycobacterium tuber- biosafety cabinet use; special floor and ceiling
culosis materials specified.
BSL 4 Dangerous and exotic agents that pose Ebola virus Hazmat suit and self-contained oxygen system, entrance
a high risk of aerosol-transmitted Marburg virus containing multiple showers, a vacuum room, an
laboratory infections, or which Smallpox ultraviolet light room, and multiple airlocked doors.
produce severe or fatal disease in Strict control of laboratory access to authorized
humans personnel.

Negative test results can also occur when the cell line inocu-
BOX 1-2 lated is not sensitive to the virus present in the specimen. Low-­
passage cells may also lose their infectivity for viral infection if
Reasons for Negative Test Results following Isolation they have undergone multiple serial passages.
of Viruses in Cell Culture False negatives can also occur if the plates are overgrown by
bacteria or fungi. Although treatment of the viral transport and
Virus not causing the disease culture media with antimicrobials can help prevent this, resis-
Virus causing the disease, but: tant bacteria or fungi may still be present. Sometimes, multiple
Specimen size inadequate viruses are present within a specimen, and one virus overgrows
No viral shedding at the time of sampling another. For example, this can occur with mixed infections with
No viral shedding at the site of sampling FCV and feline herpesvirus-1 (FHV-1) in cats with upper respi-
Antibody interference with viral infectivity for cell ratory tract disease. FCV rapidly infects Crandell-Reese feline
culture kidney cells and produces CPE, which obscures the concurrent
Inadequate organism numbers at the anatomic site presence of FHV-1.
of specimen collection
Loss of organism viability during transportation to Positive Results
the laboratory It is imperative that veterinary clinicians be aware that the
Overgrowth of one viral pathogen by another detection of a virus in a specimen does not always imply that
Overgrowth by bacterial or fungal agents the organism is the cause of the animal’s clinical signs. This is
Lack of cell sensitivity for the virus (wrong cell type especially true for specimens collected from the respiratory or
inoculated) gastrointestinal tracts, where multiple infectious agents may
Laboratory inexperience with techniques required be present concurrently, and in many cases, viruses can repli-
for virus isolation and identification cate in these locations without causing clinical signs of illness.
In some animals, the development of severe clinical signs is
more likely when there is simultaneous presence of multiple
infectious agents. As noted previously for specimens that test
inadequate organism numbers at the anatomic site of specimen negative, the presence of one agent may also obscure the pres-
collection, or loss of organism viability during transportation ence of another, more significant organism (such as with FCV
to the laboratory. Loss of viral viability is more likely to occur and FHV-1 co-infections), which could result in the incorrect
with enveloped viruses such as canine distemper virus than with assumption that only one organism is the cause of an animal’s
non-enveloped, hardy viruses such as canine parvovirus or FCV. disease.
CHAPTER 1  Isolation in Cell Culture 9

REFERENCES 17. England JJ, Poston RP. Electron microscopic identification and
subsequent isolation of a rotavirus from a dog with fatal neonatal
1. Heikkinen T, Marttila J, Salmi AA, et al. Nasal swab versus naso- diarrhea. Am J Vet Res. 1980;41(5):782-783.
pharyngeal aspirate for isolation of respiratory viruses. J Clin 18. Bao L, Xu L, Zhan L, et al. Challenge and polymorphism analysis
Microbiol. 2002;40(11):4337-4339. of the novel (H1N1) influenza virus to normal animals. Virus Res.
2. World Health Organization guidance on regulations for the Trans- 2010;151(1):60-65.
port of Infectious Substances 2009-2010. http://www.who.int/csr/ 19. Essex M, Kawakami TG, Kurata K. Continuous long-term replica-
resources/publications/biosafety/WHO_HSE_EPR_2008_10/en/ tion of feline leukemia virus (FeLV) in an established canine cell
index.html. Last accessed January 25, 2013. culture (MDCK). Proc Soc Exp Biol Med. 1972;139(1):295-299.
3. Schobesberger M, Zurbriggen A, Summerfield A, et al. Oligoden- 20. Nishikawa Y, Iwata A, Nagasawa H, et  al. Comparison of the
droglial degeneration in distemper: apoptosis or necrosis? Acta growth inhibitory effects of canine IFN-alpha, -beta, and -gamma
Neuropathol. 1999;97(3):279-287. on canine cells infected with Neospora caninum tachyzoites. J Vet
4. Maeda Y, Tohya Y, Matsuura Y, et al. Early interaction of canine Med Sci. 2001;63(4):445-458.
calicivirus with cells is the major determinant for its cell tropism 21. Crandell RA, Fabricant CG, Nelson-Rees WA. Development, char-
in vitro. Vet Microbiol. 2002;87(4):291-300. acterization, and viral susceptibility of a feline (Felis catus) renal
5. Le LP, Rivera AA, Glasgow JN, et al. Infectivity enhancement for cell line (CRFK). In Vitro. 1973;9(3):176-185.
adenoviral transduction of canine osteosarcoma cells. Gene Ther. 22. Kreutz LC, Seal BS, Mengeling WL. Early interaction of feline cali-
2006;13(5):389-399. civirus with cells in culture. Arch Virol. 1994;136(1-2):19-34.
6. Hemphill A, Vonlaufen N, Golaz JL, et  al. Infection of primary 23. Van Hamme E, Dewerchin HL, Cornelissen E, et  al. Attachment
canine duodenal epithelial cell cultures with Neospora caninum. and internalization of feline infectious peritonitis virus in feline
J Parasitol. 2009;95(2):372-380. blood monocytes and Crandell feline kidney cells. J Gen Virol.
7. Skloot R. The Immortal Life of Henrietta Lacks. New York: Crown 2007;88(Pt 9):2527-2532.
Publishers; 2010. 24. Hirasawa T, Tsujimura N, Konishi S. Multiplication of canine par-
8. Mochizuki M. Growth characteristics of canine pathogenic viruses vovirus in CRFK cells. Nippon Juigaku Zasshi. 1985;47(1):89-99.
in MDCK cells cultured in RPMI 1640 medium without animal 25. Talbott RL, Sparger EE, Lovelace KM, et al. Nucleotide sequence
protein. Vaccine. 2006;24(11):1744-1748. and genomic organization of feline immunodeficiency virus. Proc
9. Young L, Sung J, Stacey G, et al. Detection of Mycoplasma in cell Natl Acad Sci USA. 1989;86(15):5743-5747.
cultures. Nat Protoc. 2010;5(5):929-934. 26. Goodman JL, Nelson C, Vitale B, et  al. Direct cultivation of the
10. Ossiboff RJ, Sheh A, Shotton J, et  al. Feline caliciviruses (FCVs) causative agent of human granulocytic ehrlichiosis. N Engl J Med.
isolated from cats with virulent systemic disease possess in  vitro 1996;334(4):209-215.
phenotypes distinct from those of other FCV isolates. J Gen Virol. 27. Binn LN, Marchwicki RH, Stephenson EH. Establishment of a
2007;88(Pt 2):506-517. canine cell line: derivation, characterization, and viral spectrum.
11. Narang HK. Ultrastructural study of long-term canine distemper virus Am J Vet Res. 1980;41(6):855-860.
infection in tissue culture cells. Infect Immun. 1982;36(1):310-319. 28. Wills PJ, Johnson L, Thompson RG. Isolation of Chlamydia
12. Lan NT, Yamaguchi R, Uchida K, et al. Growth profiles of recent using McCoy cells and Buffalo green monkey cells. J Clin Pathol.
canine distemper isolates on Vero cells expressing canine signal- 1984;37(2):120-121.
ing lymphocyte activation molecule (SLAM). J Comp Pathol. 29. Mochizuki M, Mitsutake Y, Miyanohara Y, et  al. Antigenic and
2005;133(1):77-81. plaque variations of serotype II feline infectious peritonitis corona-
13. Cory J, Yunker CE, Ormsbee RA, et al. Plaque assay of rickettsiae viruses. J Vet Med Sci. 1997;59(4):253-258.
in a mammalian cell line. Appl Microbiol. 1974;27(6):1157-1161. 30. Horimoto T, Kasaoka T, Tuchiya K, et al. An improved method for
14. Buckley SM. Survival of Toxoplasma gondii in mosquito cell lines hemagglutinin extraction from feline herpesvirus type 1-infected
and establishment of continuous infection in Vero cell cultures. Exp cell line. Nippon Juigaku Zasshi. 1989;51(1):177-183.
Parasitol. 1973;33(1):23-26. 31. Iqbal Z, Chaichanasiriwithaya W, Rikihisa Y. Comparison of PCR
15. Cornwell HJ, Weir AR, Wright NG, et al. The susceptibility of a with other tests for early diagnosis of canine ehrlichiosis. J Clin
dog kidney cell-line (MDCK) to canine distemper, infectious canine Microbiol. 1994;32(7):1658-1662.
hepatitis and canine herpesvirus. Res Vet Sci. 1970;11(6):580-582.
16. Basak S, Compans RW. Polarized entry of canine parvovirus into
an epithelial cell line. J Virol. 1989;63(7):3164-3167.
CHAPTER 2

Immunoassays
Jane E. Sykes and Shelley C. Rankin

w KEY POINTS
• I mmunoassays are commonly used in practice to detect anti- • W  hen interpreting the results of immunoassays, veterinarians
gen or antibody in body fluids. should consider what is being measured (antigen or antibody),
• The term serology is most often used to refer to assays that the likelihood of antigen or antibody being present in relation
detect antibody in serum. to the onset of illness, whether the positive test result is consis-
• Immunoassays that detect antigen or antibody in dogs and cats tent with the animal’s clinical signs, and the animal’s vaccina-
suspected to have infectious diseases include direct and indi- tion history and immune status. The possibility of false positives
rect immunofluorescent antibody (IFA), immunohistochemistry due to immunologic cross-reactivity, as well as the analytical
(IHC), ELISA, Western immunoblotting, agglutination tests, gel sensitivity and specificity of the test used (i.e., the test’s perfor-
immunodiffusion assays, and serum neutralization. mance), must also be considered.
• Positive immunoassay results do not always imply infection, • Additional assays may be required to support immunoassay
and negative results do not rule out infection. test results.

INTRODUCTION some diseases, such as leptospirosis, a significant rise occurs within


3 to 4 days of the onset of clinical signs. In secondary, or anamnes-
Serology refers to the measurement of antigen-antibody interac- tic, immune responses, the lag phase is shorter, the antibody titer
tions for diagnostic purposes, but is most often used to refer to increases more rapidly and to a greater magnitude, and antibodies
the detection of antibodies in serum. In general, a blood sample may persist for longer periods of time (months to years).
is seroreactive (or seropositive) if it contains antibodies to a par- Serologic diagnosis of infectious diseases relies on an understand-
ticular pathogen. Assays have been designed that detect differ- ing of the timing of the antibody response in relation to development
ent classes of antibodies in serum, most commonly IgG or IgM. of clinical signs for different infectious diseases. The results of sero-
Methods used to detect antibodies can also be applied to detect logic testing are interpreted differently for acute infections than
antigen. They can also be used on body fluids other than serum, they are for chronic or persistent infections. For infections with a
such as urine, cerebrospinal fluid, and aqueous humor. Assays that short incubation period, such as leptospirosis or Rocky Mountain
measure antigen-antibody interactions for diagnostic purposes spotted fever, antibody titers are often negative in the first week of
are most broadly referred to as immunoassays. Types of immu- illness. In general, an increase in antibody titers over a 2- to 4-week
noassays include immunofluorescent antibody (IFA) tests, ELISAs, period then occurs. Seroconversion is consistent with recent infec-
agglutination tests, gel immunodiffusion tests (AGID or gel ID tion, and at least a fourfold increase in titer is required for the
tests), and Western blotting. Many of these assays involve the use change to be considered significant. Because serologic test results
of polyclonal antibodies, or, more commonly, monoclonal antibod- can differ between laboratories that perform the same assay, the
ies, which are commercially available. Polyclonal antibodies are same laboratory should be used to determine the acute and the
produced in animals (e.g., rabbits or horses) as a normal antibody convalescent titer. Ideally, to minimize interassay variation, an ali-
response to antigen exposure. Monoclonal antibodies are pro- quot of the acute specimen should be stored, frozen, and assayed
duced in almost unlimited amounts in tissue culture, and have a at the same time as the convalescent specimen, although in reality
higher degree of antigenic specificity. this is often not done. A fourfold decline in antibody titer may also
Some immunoassays, such as ELISA and IFA, provide quantita- be consistent with recent infection, depending on the stage of ill-
tive results, expressed as optical density units (ODs) or titers (e.g., ness at which serologic testing is performed. For chronic, persistent
1:800, or reciprocal titer = 800). Exposure to an infectious agent infections, such as FIV infection or canine leishmaniosis, a single,
results in a rise in antibody titer over time (Figure 2-1). Usually, the positive antibody titer can indicate active infection. Titer increases
first antibody class to be produced by plasma cells is IgM, after do not occur when infection persists beyond 1 to 2 months, and so
which IgG synthesis by plasma cells occurs. When exposure to a acute- and convalescent-phase serology is not useful for diagnosis
pathogen occurs for the first time, there is a lag phase of 5 to 7 days of chronic infections.
before IgM antibodies can be detected in the blood (primary anti-
body response). The antibody titer increases, and if the infectious Specimen Collection and Transport
agent is cleared, there is a progressive decline in the titer over weeks
to months. The speed and magnitude by which the antibody titer Blood collected for antibody testing should be allowed to clot,
rises depends on host factors and the infectious agent involved. For and the serum separated immediately and then refrigerated or

10
CHAPTER 2  Immunoassays 11

FIGURE 2-1  Time course of primary and secondary immune responses, showing rela- FIGURE 2-2  Direct fluorescent antibody stain of brain tissue for rabies virus.
tive amounts of IgM and IgG produced at different times after antigen exposure. (Modified Frozen tissue sections were stained with a commercial reagent that contains three mono-
from Tizard IR. Veterinary Immunology, 8 ed. St Louis: Saunders; 2008) clonal antibodies specific for the rabies virus nucleocapsid. Antibodies were labeled with
fluorescein. Positive staining is present in a Purkinje cell. (Courtesy of Galligan J. New York
State Department of Health. In Maclachlan NJ, Dubovi EJ. Fenner’s Veterinary Virology, 4
frozen. Specimens can be refrigerated for up to a week before ed. New York: Elsevier; 2011.)
they are shipped to the laboratory. If testing is to be delayed for
longer than a week, specimens should be frozen at −20°C or
−80°C. Specimens can be stored frozen for years without loss of
antibody titer. For specimens collected for antigen testing, assay
results may be more susceptible to variation with specimen stor-
age. The laboratory should be contacted in order to determine
specimen storage and handling requirements.

Diagnostic Methods
Immunofluorescent and Immunoperoxidase
Antibody Assays
Direct IFA (also known as direct FA) detects antigen in clinical
specimens. IFA involves the use of an antibody that has been
tagged (conjugated) with a fluorescent label (such as fluores-
cein isothiocyanate) to detect the presence a specific antigen.
IFA is performed directly on a glass slide, and fluorescence is A
detected by microscopy. The sensitivity of direct IFA is too
low to detect individual virus particles or soluble antigen, so
usually IFA detects antigen present in association with eukary-
otic or bacterial cells. If the antigen is intracellular, pretreat-
ment of the slide to permeabilize the cells may be necessary.
The glass slide is then incubated with a solution that contains
the fluorescent antibody, then washed to remove unbound
antibody, and the cells are examined using a fluorescence
microscope (Figure 2-2). Examples of the use of direct IFA
in veterinary medicine include detection of Giardia oocysts,
FeLV within monocytes in peripheral blood or bone marrow,
or canine distemper virus within epithelial cells from a con-
junctival scraping. Nonspecific fluorescence can result in false
positives using these methods, but this can be overcome with
the inclusion of proper controls and technical expertise. An B
alternative to the use of a fluorescein-conjugated antibody is
the use of an antibody that has been conjugated to an enzyme FIGURE 2-3  Immunohistochemistry. A, Hematoxylin and eosin stained section of a
such as horseradish peroxidase (immunoperoxidase). A sub- lingual ulcer from a cat infected with feline herpesvirus-1. B, The brown stained cells lining
the ulcer are positive using an immunohistochemical stain for feline herpesvirus-1. (Cour-
strate, most commonly 3,3′-diaminobenzidine (DAB), is then
tesy of Dr. Patricia Pesavento, School of Veterinary Medicine, University of California, Davis.)
added to the slides. In the presence of hydrogen peroxide, DAB
is converted to an insoluble brown precipitate, which can be Indirect IFA (also known as IFA testing, or IFAT) is generally
visualized using conventional light microscopy. This technique used to establish a serum antibody titer to an infectious agent,
is known as immunocytochemistry. The same method, when but it can also be used to detect antigen. When used to detect
applied to tissue sections, is known as immunohistochemistry antigen, it is more sensitive than direct IFA. For measurement of
(IHC) (Figure 2-3). antibody titers, patient serum is serially diluted in the laboratory
12 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

FIGURE 2-4  Flow-through immunoassay device (IDEXX Laboratories, Inc.). Specific antigen (for example, a recombinant protein) or antibody is immobilized on a membrane in the
device in a spot or line. 1, Patient serum that may or may not contain target antigen or antibody is mixed with a solution that contains conjugated antibody or antigen. 2-3, The solution
is allowed to flow through the device, where it becomes captured on the membrane. 4, The device is activated (snapped), which releases wash and substrate reagents within the device. 
A positive reaction appears as a colored bar, or in this case, a dot. Color development in designated spots on the membrane can indicate the presence of antibody to Anaplasma spp., Ehrlichia
canis, and Borrelia burgdorferi, or the antigen of Dirofilaria immitis.

and reacted with a known antigen that has been fixed to glass wells of the plate. The antigen is either bound directly to the
slides. IFA slides are commercially manufactured for this pur- surface of the plate or is bound through the use of a capture
pose. Slides are then washed to remove unbound antibody. A antibody, which is an antibody that is bound to the plate sur-
secondary antibody, which is designed to react with the bound face that specifically binds to the antigen of interest (sandwich
patient antibodies (e.g., dog IgG or dog IgM), is applied to the ELISA). The antigen is then detected with an antibody that is
slides, which are then washed again. This secondary antibody conjugated to an enzyme such as immunoperoxidase, and the
has been conjugated to a fluorescent label. The slides are then substrate is added. This results in a color change, which can be
examined using a fluorescence microscope. The highest dilution read visually or using an ELISA plate reader (Figure 2-5). The
of serum that results in specific fluorescence is reported to the plate reader records optical density of the wells in comparison
clinician as the antibody titer to the infectious agent of interest. to that of control wells, allowing assessment of the quantity of
Examples of the use of indirect IFA for serologic testing in dogs antigen present. The plates are washed with a detergent solution
and cats include quantitative serology for some tick-borne infec- between steps, and this can be automated using an ELISA plate
tious diseases (e.g., Ehrlichia canis, Anaplasma spp.). washer. Sandwich ELISAs have been used widely for detection
More recently, veterinary immunofluorescence assays have of FeLV antigen in cat blood. In competitive ELISA, the test
been marketed that allow multiple antigen (or antibody) targets specimen (e.g., dog serum), which contains an unknown amount
to be mounted on novel platforms, such as fluorescent beads of antigen, is mixed with a known amount of labeled antigen.
or wells on the surface of a spinning silicon disc that resem- The mixture is added to wells that contain known amounts of a
bles a compact disc. Automated silicon disc–based assays allow capture antibody, and the labeled and unlabeled antigens com-
rapid and simultaneous analysis of hundreds of specimens, with pete for antibody binding. The more labeled antigen that binds,
dramatic reduction in labor associated with traditional IFA or the less antigen is present in the test specimen. Other variations
ELISA assays, and reduced chance of false-positive or -negative of competitive ELISAs also exist.
assay results due to human error. A bead-based assay is cur- For antibody detection, serial dilutions of patient serum can
rently available in the United States for detection of antibody be added to a known amount of antigen fixed on a plate and
to Borrelia burgdorferi (see Chapter 51). A silicon disc–based detected using a secondary antibody.
assay is available in the United States for detection of antibodies
to B. burgdorferi, E. canis, and Anaplasma phagocytophilum Western Immunoblotting
and antigen to Dirofilaria immitis (Accuplex 4, Antech Diag- Western immunoblotting, also known as Western blotting, is
nostics, Irvine, Calif). The performance of these assays in the also used to detect antibodies to an infectious agent in patient
field is under investigation. serum. After physical and/or chemical disruption of an organ-
ism, the proteins are then separated on the basis of molecular
Enzyme Linked Immunosorbent Assays mass using vertical sodium dodecyl sulfate–polyacrylamide gel
ELISA (also known as enzyme immunoassay) can be used to electrophoresis (SDS-PAGE). The protein bands are then trans-
detect either antigen or antibody. In the laboratory, it is usually ferred (blotted) to a nitrocellulose membrane using an electric
performed in a polystyrene microtiter plate. However, in-clinic current. The membrane is cut into vertical strips, and each
modifications of the test, such as flow-through or lateral-flow strip is probed with serial dilutions of patient serum. If there
ELISAs (e.g., SNAP, IDEXX Laboratories, Inc.), whereby anti- are antibodies in the serum to antigens from the organism of
gen or capture antibody is immobilized on a membrane filter, are interest, the antibodies bind to the antigens on the strip. The
widely used in veterinary practices (e.g., for diagnosis of feline antigen-antibody complexes can then be visualized as discrete
retroviral, heartworm, Giardia, Leishmania, and tick-borne bands using immunoperoxidase detection methods. The pattern
infections) (Figure 2-4). For antigen detection, an unknown of band reactivity is specific for an immune response to that
quantity of antigen in a clinical specimen is immobilized in the organism (Figure 2-6). Western blotting is offered commercially
CHAPTER 2  Immunoassays 13

FIGURE 2-5  Sandwich ELISA. An antibody is immobilized in the plate, which binds antigen in a test specimen. The antigen is then detected using an antibody that is conjugated to an
enzyme such as immunoperoxidase, and the substrate is added, which results in a color change. (Modified from Tizard IR. Veterinary Immunology, 8 ed. St Louis: Saunders; 2008.)

FIGURE 2-6  Western blotting (immunoblotting). Purified viruses or bacteria are disrupted and subjected to electrophoresis along the length of a polyacrylamide gel, so that the
proteins are separated according to their molecular weight (A). The bands are then transferred to a nitrocellulose paper membrane (B) and probed with patient serum (C). The strip shown
on the right is a Western blot that shows antibody reactivity to the antigens of FIV (D). (Original blot courtesy of IDEXX Laboratories, Inc.)

for the diagnosis of FIV and B. burgdorferi infection and has latex particles. They rely on the bivalent nature of antibodies,
been used in research laboratories as the gold standard for the which can cross-link particulate antigens. Serial dilutions of
detection of antibody responses to vector-borne pathogens such serum are tested for their ability to cause or inhibit agglutination,
as E. canis. In the case of FIV infection, Western blotting has and the highest dilution that causes or inhibits agglutination is
been used to confirm positive ELISA test results.1 In the case of reported as the antibody or antigen titer. IgM causes aggluti-
B. burgdorferi infection, Western blotting has been evaluated nation more effectively than IgG. If excess antibody is present,
for its ability to differentiate between the response to vaccina- particles may become so coated with antibody that agglutina-
tion and natural exposure.2 tion is actually inhibited. This is called the prozone effect and
can result in false-negative test results if serum is not adequately
Agglutination Testing diluted. Examples of agglutination tests that are used to diag-
Agglutination tests detect antibody or antigen and involve agglu- nose infectious diseases of dogs and cats include the microscopic
tination of bacteria, red cells, or antigen- or antibody-coated agglutination test (MAT) for serologic diagnosis of leptospirosis
14 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

Agar Gel Immunodiffusion


For agar gel immunodiffusion (AGID, gel ID or double diffu-
sion) tests, round wells are cut in a layer of agar on a plastic plate
using a punch. One well, usually the central well, is filled with a
soluble antigen, and the other wells are filled with patient serum.
A positive antibody control specimen is included in each assay.
The reagents are allowed to diffuse into the agar, and when anti-
bodies and antigen meet in optimal proportions (the point of
equivalence), immune complexes form. This results in a visible
line of precipitate between the antigen well and wells that contain
positive serum (Figure 2-7). Immunodiffusion testing is most often
used to detect antibodies to fungal pathogens in dogs, such as
Aspergillus fumigatus,3 Coccidioides immitis,4 and Blastomyces
dermatitidis.5 It is also used to detect antibodies to Brucella canis.

Serum Neutralization
Serum neutralization is a technique used to determine the anti-
body titer to viruses. Serial dilutions of patient serum are mixed
with a known quantity of virus particles. The mixture is incu-
bated to allow the antibodies in the patient serum to bind to the
virus particles, and the mixture is then added to a confluent cell
monolayer. The highest dilution of patient serum that inhibits
viral replication in the monolayer (as determined by examination
A for cytopathic effect or immunostaining) is the antibody titer
reported by the laboratory. Serum neutralization can be used to
detect antibodies to canine distemper virus and several respira-
tory viruses that infect dogs and cats.

Complement Fixation Testing


Complement fixation (CF) tests have largely been superseded by
the use of ELISA tests. The CF test detects antibodies in patient
serum. The serum is heat-treated to inactivate complement in the
test specimen. The test uses sheep RBCs, anti–sheep RBC anti-
body, and unheated guinea pig serum as a source of complement.
Under normal circumstances, when these three reagents are mixed,
the result is hemolysis. Hemolysis occurs when complement binds
to antibody-coated RBC and triggers formation of the membrane
attack complex, with resultant pore formation. In the CF test, a
known antigen is added to the heat-treated patient serum. When
B antibodies are present in the serum, the addition of this antigen
leads to the formation of antigen-antibody complexes. Guinea pig
FIGURE 2-7  A, Gel immunodiffusion (or double diffusion) system used for detection of complement is then added to the mixture and is consumed by the
antibodies to Aspergillus fumigatus, Histoplasma capsulatum, Blastomyces dermatitidis, and
complexes. The resultant mixture is thus unable to lyse sensitized
Coccidioides immitis (Meridian Bioscience, Inc.). The kit consists of an agar diffusion medium
in a small plastic tray that contains four arrays of six wells arranged around a central well.  sheep RBCs. A lack of hemolysis indicates a positive CF test.
An antigen preparation for each of the fungal species is placed in the central well of each
array. B, Positive gel immunodiffusion for Aspergillus fumigatus. The central well (Ag) Interpretation of Results
contains a purified carbohydrate preparation from mycelial phase cultures of Aspergillus
fumigatus, Aspergillus niger, and Aspergillus flavus. The top two wells (D1) and the bottom To interpret the results of serology or immunoassay tests, the
two wells (D2) contain serum from 2 dogs (dog 1 and dog 2) with nasal aspergillosis (per- clinician first needs to consider whether the test was designed to
formed in duplicate for each dog). After the wells are loaded, the plate is incubated at room detect antibody or antigen (Tables 2-1 and 2-2), and, if designed
temperature for 24 hours. Precipitin lines form when the antigen and antibody diffuse out to detect antibody, whether it was IgM-specific (detects early
of each well and form immune complexes. A light box is used to illuminate the plate so the antibody responses). The results also have to be interpreted in
precipitin bands are visible. Both dogs are positive for antibody to the Aspergillus species
light of the pathogenesis of the infectious disease, specifically
tested (small arrows), although the reaction is strongest for dog 1. The arrowheads are the
precipitin lines generated by the positive control antibodies (in wells labeled C). the timing of antigen and antibody appearance in the body fluid
being tested. Finally, the sensitivity and specificity of the test
itself need to be considered (i.e., the analytical sensitivity and
(agglutination of live leptospires) and the cryptococcal antigen specificity), which are determined during test validation (Table
latex agglutination test (agglutination of antibody-coated latex 2-3). For example, some immunoassays that detect antigen will
beads). Hemagglutination inhibition (HI) is used to determine not generate a positive result unless a large amount of antigen
antibody titers to canine parvovirus and canine influenza virus, is present in the specimen provided (low analytical sensitivity).
and it evaluates the ability of serum to inhibit erythrocyte agglu- Test analytical sensitivity and specificity should be available
tination by these viruses. from the laboratory that performs the test. False-positive test
CHAPTER 2  Immunoassays 15

TABLE 2-1
Guidelines for Interpretation of Test Results Using Immunoassays That Detect Antigen
Antigen Test Result Possible Explanation Troubleshooting Suggestions
Negative Pathogen of interest not present Confirm with additional diagnostic test if necessary, (e.g.,
alternative antigen assay, antibody or PCR assay)
Antigen present, but quantity below Consider an antibody or PCR assay
limit of detection
Antigen present but complexed by Consider an antibody or PCR assay
antibody and not available for
detection
Technical error or reagent failure Identified with inclusion of appropriate laboratory controls
Positive Pathogen of interest present Confirm with additional diagnostic test if necessary, especially
in regions with a low prevalence of infection. Antigen pres-
ence may not imply that the pathogen is the cause of disease
Antigen present but viable microbe Confirm with additional diagnostic test
absent
Vaccine antigen present Obtain vaccination history and consider a test that discrimi-
nates between vaccine and wild-type organisms, if available
Cross-reactive antigen present Obtain history and consider the likelihood of infection with
from another substance or related agents
pathogen
Technical error Identified with inclusion of appropriate controls by the
­laboratory
Poor assay analytical specificity Use alternative assay
(prone to generate false-positives)

TABLE 2-2
Guidelines for Interpretation of Test Results Using Immunoassays That Detect Antibody
Antibody Test Result Possible Explanation Troubleshooting Suggestions
Negative Exposure to pathogen of interest Confirm with additional diagnostic test if necessary
did not occur
Too early in course of infection/ Obtain convalescent titer; consider an antigen or PCR test for
illness early diagnosis
Severe immunosuppression Consider an antigen or PCR test
Technical error Identified with inclusion of appropriate controls by the
­laboratory
Poor assay analytical sensitivity Use alternative assay
(prone to false-negatives)
Positive Previous natural exposure to the If infection is chronic and persistent, positive test results may
pathogen of interest equal active infection.
Previous immunization against Obtain vaccination history; consider use of assays that dis-
the pathogen of interest criminate between natural exposure and vaccine antibody, if
available
Immunologic cross-reactivity Obtain history and consider the likelihood of previous exposure
to related agents
Technical error Identified with inclusion of appropriate controls by the
­laboratory
Poor assay specificity (prone to Use alternative assay. Consider the use of a confirmatory test,
­false-positives) especially in regions with low disease prevalence
16 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

that the pathogen is the cause of an animal’s clinical signs. Anti-


TABLE 2-3 gen can also be present in the absence of intact, viable organisms.
Sensitivity, Specificity, Positive and Negative Predictive Value Positive results can follow recent vaccination with the pathogen
of interest. For example, positive fecal parvovirus antigen tests
Test Attribute Definition can occasionally occur in fecal specimens obtained several days
after attenuated live virus vaccination of kittens for feline panleu-
Sensitivity Number of truly positive animals that are kopenia virus.6 False positives can occur as a result of antigenic
identified as positive by the test, divided cross-reactivity with some other substance or organism in the
by the total number of animals that are specimen. An example is positive serum and urine antigen assays
truly positive (as determined by the gold for Aspergillus spp. in dogs treated with Plasmalyte infusions or
standard assay) (%). A test with low those infected with other molds such as Paecilomyces spp.7
sensitivity fails to detect many positive
animals (many false-negative results). Negative Results for Antibody Detection
Specificity Number of truly negative animals that are Negative results for antibody will occur if the patient has not
identified as negative by the test, divided by been exposed to the infectious agent of interest. They may also
the total number of animals that are truly occur if the patient has not yet had time to develop antibod-
negative (as determined by the gold stan- ies. For example, negative antibody tests early in the course of
dard assay) (%). A test with low specificity illness are common with leptospirosis and canine granulocytic
generates many false-positive test results. anaplasmosis. In this situation, a convalescent titer is required
Positive pre­ The number of animals that are truly for diagnosis using serology. Negative antibody test results
dictive value positive divided by the total number of can occur in severely immunosuppressed animals, such as in
animals that test positive (true positives cats with advanced FIV infection that no longer can mount an
plus false positives). immune response, or cats with advanced feline infectious peri-
tonitis. Negative results may also occur because the test lacks
Negative pre­ The number of animals that are truly
sensitivity (i.e., low analytical sensitivity).
dictive value negative divided by the total number of
animals that test negative (true negatives Positive Results for Antibody Detection
plus false negatives).
Positive results for IgG antibody suggest previous exposure
Etiologic pre­ The proportion of animals with positive test to a pathogen and do not necessarily indicate active infection.
dictive value* results that truly have the disease of interest, Paired IgG titers are required to document recent infection.
as opposed to carriage of an organism in Single positive results for IgM-specific assays may indicate a
the absence of disease. recent infection or a chronic infection that continues to stimu-
late the immune system. Previous immunization may also result
*Gunnarson, RK, Lanke J. The predictive value of microbiologic
in positive results. If the infection is chronic and persistent, such
diagnostic tests if asymptomatic carriers are present. Statist Med 2002;
21:1773–1785. as with FIV infection, positive results indicate active infection,
provided there has been no history of previous FIV vaccina-
tion. False-positive results for antibody may also occur when
results due to low analytical specificity are more likely when antigenic cross-reactivity occurs. For example, dogs recovering
testing animals from populations where the prevalence of true from Leptospira infection have been shown to test positive on
positive test results is low. In this situation, a test has a low posi- IFA for the related spirochete B. burgdorferi.
tive predictive value, and confirmatory testing may be required.
A quality assurance program should be in effect within the labo- REFERENCES
ratory to ensure that specimens submitted are of adequate qual- 1. Levy J, Crawford C, Hartmann K, et al. 2008 American Association
ity, that sufficient technical expertise is present to perform the of Feline Practitioners’ feline retrovirus management guidelines.
tests, and that appropriate control assays are included with each J Feline Med Surg. 2008;10(3):300-316.
run to ensure proper assay performance. 2. Leschnik MW, Kirtz G, Khanakah G, et  al. Humoral immune
response in dogs naturally infected with Borrelia burgdorferi sensu
Negative Results for Antigen Detection lato and in dogs after immunization with a Borrelia vaccine. Clin
Vaccine Immunol. 2010;17(5):828-835.
Negative results for antigen will occur if the antigen is not present 3. Pomrantz JS, Johnson LR, Nelson RW, et al. Comparison of sero-
in the specimen being tested. They may also occur if the amount logic evaluation via agar gel immunodiffusion and fungal culture
of antigen is below the limit of detection of the assay (e.g., of tissue for diagnosis of nasal aspergillosis in dogs. J Am Vet Med
low worm burden in heartworm disease). For some infectious Assoc. 2007;230(9):1319-1323.
agents, such as FIV, antigen testing is not performed because 4. Pappagianis D, Zimmer BL. Serology of coccidioidomycosis. Clin
antigen levels during infection are typically extremely low. The Microbiol Rev. 1990;3(3):247-268.
amount of antigen present can also vary over the course of a 5. Spector D, Legendre AM, Wheat J, et  al. Antigen and antibody
particular infection. Finally, false-negative results may occur testing for the diagnosis of blastomycosis in dogs. J Vet Intern
Med. 2008;22(4):839-843.
when antibody present in the specimen complexes antigen and
6. Patterson EV, Reese MJ, Tucker SJ, et  al. Effect of vaccina-
makes it unavailable for detection by the immunoassay. tion on parvovirus antigen testing in kittens. J Am Vet Med Assoc.
2007;230(3):359-363.
Positive Results for Antigen Detection 7. Garcia RS, Wheat LJ, Cook AK, et al. Sensitivity and specificity of
Positive results for antigen can indicate the presence of the sus- a blood and urine galactomannan antigen assay for diagnosis of
pected pathogen in the specimen. They do not necessarily imply systemic aspergillosis in dogs. J Vet Intern Med. 2012;26:911-919.
CHAPTER 3
Isolation and Identification of Aerobic
and Anaerobic Bacteria
Jane E. Sykes and Shelley C. Rankin

w KEY POINTS
• B  ecause of the increasing prevalence of antimicrobial drug • C  oncurrent examination of a Gram-stained smear by the labo-
resistance among bacteria isolated from dogs and cats, culture ratory assists in assessment of specimen quality and interpreta-
and susceptibility is always recommended when a bacterial tion of culture results.
infection is suspected at a normally sterile anatomic site. • Antimicrobial susceptibility testing is most commonly per-
• Successful isolation of aerobic and anaerobic bacteria from a formed using broth microdilution or agar diffusion testing and
clinical specimen relies on collection of a sufficient quantity aims to determine the minimum concentration of an antimi-
of material, from the correct location, and appropriate storage crobial drug that inhibits growth of the organism in vitro (mini-
and transport of the specimen to the laboratory. Care should be mum inhibitory concentration, or MIC). The laboratory uses
taken to prevent specimen contamination during collection. MIC breakpoints, which are established and revised regularly
• The laboratory should be provided with an appropriate history by national standards agencies, to assess the susceptibility of
if fastidious, anaerobic, or slow-growing bacterial organisms an organism to a particular antimicrobial drug.
are suspected.

INTRODUCTION Collection of a sufficient amount of specimen is very important.


Table 3-1 suggests optimal specimen types for detection of bac-
Factors that influence the detection of clinically relevant organ- teria from specific anatomic sites. Attempts should always be
isms in specimens collected from dogs and cats are: made to prevent contamination with commensal bacteria during
1. An appropriate level of suspicion for the presence of a bacterial specimen collection. When anaerobes are suspected, aspirates or
infection; biopsies should be obtained rather than swabs, and the speci-
2. Development of a list of differential diagnoses that reflects men should be stored at room temperature and not refrigerated.
the types of bacterial pathogens that might be the cause of This is because oxygen diffuses into cold specimens more rap-
clinical signs, because of the requirement of some bacteria for idly than specimens held at room temperature.
special transport conditions, culture conditions, or prolonged Specimens should be transported to the laboratory imme-
incubation; diately. As a general rule for aerobic culture, if transport
3. Collection of a sufficient amount of specimen; requires longer than 2 hours, appropriate transport medium
4. Collection of the correct type of specimen; should be used or specimens should be refrigerated without
5. Specimen collection from the correct anatomic location; and transport medium. Although specimens can be held for up to
6. Proper handling, storage and transport of the specimen, which 72 hours, culture of specimens older than 24 hours should
should be sent with a request for culture to the laboratory be avoided, even if they have been refrigerated or placed
within acceptable time limits. in transport media, because of increased likelihood of false
These factors cannot be overemphasized. Without them, time and positive and false negative results with longer storage times.
money invested in collection of specimens and culture is wasted. Transport medium for bacterial culture is usually supplied in
Given the increasing prevalence of multidrug resistance among a plastic tube with one or two swabs attached to the tube’s
bacteria that infect dogs and cats, veterinarians should make cap (Figure 3-1). The medium is designed to maintain bac-
attempts to confirm a bacterial infection at a normally sterile site terial viability without causing significant growth. It usually
by requesting microscopic evaluation of direct smears and culture consists of a small amount of agar, reagents that maintain pH,
by a laboratory whenever possible before the choice is made to a colorimetric pH indicator that indicates whether oxidation
administer an antimicrobial drug. has occurred, and specific factors that maintain the viability
The purpose of this chapter is to provide information in regard of certain pathogens. Some organisms, such as chlamydiae
to the correct way to collect and handle specimens for culture and leptospires, have specific transport medium requirements.
before they are sent to the laboratory, and to outline bacteriologic Specimens that are placed in transport media should not be
procedures performed in the microbiology laboratory that are of refrigerated.
relevance to the small animal clinician. Specimens should be labeled with the patient’s name, patient
number, source of specimen, and the date and time of collection,
Specimen Collection and Transport and should be labeled, packaged, and transported according to
regional and national regulations (see Chapter 1). Specimens
Veterinary clinicians should collect specimens for culture for anaerobic culture should preferably be transported in an
using an appropriate method from the correct anatomic site. oxygen-free container.

17
18 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

TABLE 3-1
Suggested Specimen Types for Bacterial Culture from Selected Anatomic Sites and Likely Pathogens Present
Specimen Type Suggested Volume for Culture Likely Pathogens Present
Blood ≥10 mL for patients weighing >10 kg, or 1% Gram-positive and gram-negative aerobes. Common contam-
of patient blood volume inants are coagulase-negative staphylococci, Corynebacte­
rium, Bacillus spp., and propionibacteria (see Chapter 86)
CSF ≥0.5 mL Gram-positive and gram-negative aerobes, anaerobes,
­Brucella spp. (dogs), Nocardia, Actinomyces,1 Myco­
plasma spp. (cats)2 (see Chapter 90)
Middle ear Aspirate of fluid or pus through tympanic Gram-positive and gram-negative aerobes, anaerobes,
membrane ­Mycoplasma spp., Actinomyces spp.2,3 (see Chapter 84)
Skin Swab from epidermal collarette or ruptured pustule Gram-positive aerobes (especially staphylococci), rarely
(superficial pyoderma); biopsy from which dermis Pseudomonas aeruginosa (see Chapter 84)
has been removed (deep pyoderma)
External ear Swab ear canal after removing debris and crusts Gram-positive aerobes, Pseudomonas aeruginosa (see
from the canal Chapter 84)
Cornea Scrape or swab exposed corneal stroma and inocu- Gram-positive aerobes, Mycoplasma spp., Pseudomonas
late directly onto media. Referral to veterinary aeruginosa4
ophthalmologist recommended
Abdominal fluid 1-5 mL Gram-negative and gram-positive aerobes, anaerobes,
Actinomyces spp. (see Chapter 88)
Pleural fluid 1-5 mL Gram-negative and gram-positive aerobes, anaerobes,
Actinomyces spp., Nocardia spp., Mycoplasma spp.
(see Chapter 87)
Feces Fresh whole stools preferred. Avoid swabs or fecal Escherichia coli, Salmonella, Clostridium perfringens,
loops containing fecal matter due to low specimen Clostridium difficile, Campylobacter spp., Anaerobio­
size. Submit in clean, dry leak-proof container. spirillum (Chapters 45-49 and 88)
Urine 1-5 mL urine collected using cystocentesis Gram-negative and gram-positive aerobes, Coryne­bacterium
urealyticum (see Chapter 89)
Bronchoalveolar 1-5 mL Gram-positive and gram-negative aerobes, anaerobes
lavage (aspiration pneumonia), Mycoplasma spp., rarely Myco­
specimens bacterium spp. (see Chapter 87)

results of culture and susceptibility testing. If sufficient material


is available, examination of a direct smear also helps to deter-
mine whether a specimen is adequate for culture and aids inter-
pretation of culture results. For swab or aspirate specimens,
clinicians should consider providing a separate specimen for a
direct smear in addition to a specimen for culture.
Acid-fast stains can be used on smears to assist in the identifi-
cation of Nocardia and Mycobacterium spp., as well as on fecal
smears to detect some parasitic oocysts, such as those of Cryp­
tosporidium. These organisms stain magenta as a result of their
FIGURE 3-1  Swab system containing transport media for aerobic bacterial culture. high cell wall mycolic acid content, which in turn results in their
resistance to decolorization after staining (which is often per-
formed using an acid-alcohol solvent, hence the term acid-fast).
Diagnostic Methods Examples of acid-fast staining methods are the Ziehl-Neelsen,
Fite’s, and Kinyoun stains.
Microscopic Examination of Direct Smears Other microscopic techniques used for bacterial identifica-
A Gram stain prepared from the specimen can permit the rapid tion in specialized laboratories include darkfield microscopy,
preliminary diagnosis of infection and determine whether the phase-contrast microscopy, and fluorescence microscopy (see
organism(s) present are gram-positive or gram-negative (Box Chapter 2). Darkfield and phase contrast microscopy allow visu-
3-1; Figure 3-2). This helps guide the clinician to select an alization of organisms without fixation and staining, so motility
appropriate empiric therapy, if necessary, while awaiting the as well as structure can be assessed. In darkfield microscopy, the
CHAPTER 3  Isolation and Identification of Aerobic and Anaerobic Bacteria 19

condenser only permits light that bounces off an object to pass


BOX 3-1 from below up into the objective, so the object appears bright
against a dark background. Darkfield microscopy is used pri-
Procedure for Gram Staining marily for identification of fine spirochetes such as leptospires,
which cannot be visualized using conventional microscopy (see
Heat-fix specimen on the slide over a gentle flame or fix in Chapter 50). In phase-contrast microscopy, slowing and deflec-
95% methanol for 2 min, and then air dry tion of beams of light occur as they pass through an object.
Flood with crystal violet solution for 15 to 60 s (10 g of
90% dye in 500 mL absolute methanol) Bacterial Culture
Wash with water The performance of bacterial culture for diagnostic purposes
Flood with iodine for 15 to 60 s (6 g of I2 and 12 g of KI requires appropriate laboratory facilities, proper biosafety
in 1800 mL of water) level-2 containment, properly trained individuals, reference
Decolorize with acetone-alcohol (400 mL of acetone in strains for quality control testing, and the use of standard pro-
1200 mL of 95% ethanol) tocols.5 Media used for bacterial culture may be categorized as
Wash immediately general-purpose, enriched, selective, differential, and specialized
Counterstain with safranin for 15 to 60 s (10 g of dye in media (Table 3-2). Some media belong to more than one of these
1L of water) categories. For example, MacConkey agar is both a selective and
Wash, blot dry, and examine using light microscopy a differential medium (Figure 3-3). Media for culture contains a
nutrient source, most commonly peptones (hydrolyzed animal
or vegetable substances), and is adjusted to a specific pH. Solid
medium contains agar, which is derived from seaweed. Other
nutrients may be added based on specific organism requirements.
More than 100 different types of bacteriology media exist, most
of which can be purchased as preprepared plates or broth.
Blood culture media contain the anticoagulant sodium poly-
anethol sulfonate (SPS), which is less inhibitory to bacterial
growth than other anticoagulants. Some blood culture media
(e.g., BD BACTEC Plus) contain resin beads that bind antimicro-
bial drugs in the patient’s blood specimen, so they do not inhibit
bacterial growth (Figure 3-4). The detection of bacterial growth
in blood culture systems can be automated using instrumentation
that measures emission of fluorescence or a colorimetric change
in a sensor. The change occurs as CO2 concentrations within
the bottle increase, which indicates the presence of bacterial
growth. Some instruments detect pressure changes in the head
of the bottle. The culture is monitored at regular intervals by
the instrument, to optimize early detection of bacterial growth.
Positive blood culture bottles are then evaluated by performing
FIGURE 3-2  Gram-stained smear of gram-positive cocci (dark blue) and gram-­
negative rods (red) (1600× oil magnification). a Gram stain of the broth, and subculture to appropriate media.

TABLE 3-2
Types of Media Used in Bacteriology
Medium Type Attributes Example
Transport Nonnutritive liquid or semisolid medium. Promotes Amies transport medium
­organism viability without allowing significant
growth.
General-purpose Allows growth of most aerobic and facultatively Sheep blood agar
­anaerobic organisms
Enriched Promotes growth of fastidious organisms that require Chocolate agar, which is enriched with heat-
specific nutrients, for example Francisella treated blood, which gives it a brown color
Selective Selects for growth of certain organisms while inhibiting MacConkey agar, which selects for gram-­
others negative bacteria
Differential Aids in preliminary identification of organisms based MacConkey agar, which promotes pink colony
on colony appearance formation by organisms that ferment lactose
Specialized Selects for a specific pathogen through the addition of Anaerobic medium, which contains hemin,
specific nutrients vitamin K, and reducing agents
20 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

Another blood culture system is the Isolator lysis centrifuga- by providing an adequate clinical history, when pathogens with
tion system (DuPont, Wilmington, DE) (See Figure 3-4, B). In special culture requirements are suspected, such as Campylo­
this system, blood is added to a tube that contains anticoagulant bacter, Mycoplasma, Actinomyces, Nocardia, and Mycobacte­
and a reagent that induces RBC lysis. The tube is centrifuged, rium spp. These organisms require special media or prolonged
and the pellet is plated onto agar media and incubated. This incubation times (e.g., weeks). Specimens for anaerobic culture
method allows colony counts to be obtained. The tubes allow should be plated onto appropriate media immediately and
isolation not only of aerobes, but also some anaerobic bacteria. incubated in an anaerobic environment. Anaerobic containers,
In the laboratory, media for bacterial culture should be which create an anaerobic environment through the use of gas-
warmed to room temperature before inoculation, and dam- generating reagents, can be used for incubation (Figure 3-5).
aged media, such as dehydrated medium or medium that has Because anaerobes grow more slowly than aerobes, anaerobic
changed color, should not be used. Medium is stored in the dark cultures should be held for 7 days before being reported as
at 2°C to 8°C and discarded when it reaches its expiration date. negative.
After plating, incubation is most commonly performed at 35°C
to 37°C in air for a minimum of 2 days. Conditions vary for Blood and Intravascular Catheters
specific bacteria, and growth in 3% to 5% CO2 is preferred by Bacteremic animals may be continuously or intermittently bac-
some fastidious organisms. The laboratory should be informed, teremic. Because of intermittent bacteremia, multiple, separate
specimens, each containing greater than or equal to 10 mL of
blood, are recommended. Pediatric bottles are available that
can be inoculated with as little as 1.5 mL of blood, although
the chance of a positive isolation increases as the blood volume
drawn increases. Therefore, as much blood should be drawn as
possible, provided that collection of a large volume of blood
does not jeopardize the patient. Bottles should be held at room
temperature and submitted immediately to the laboratory.
Two, and ideally three, blood samples should be collected using
meticulous sterile technique, and the timing of blood draws
should depend on the severity of the animal’s illness (Box 3-2).
Inoculated bottles are incubated in aerobic conditions for 7 days
before being reported as negative. Additional blood samples are
not recommended, because they add cost without a significant
increase in sensitivity.6 The choice to inoculate anaerobic blood
culture bottles is controversial. Studies in humans have shown
that the prevalence of anaerobic bloodstream infections has
decreased, and routine inoculation of anaerobic blood culture
bottles may not be necessary. In one veterinary study, anaerobes
were not identified in any of 33 dogs with endocarditis using
FIGURE 3-3  Growth of bacteria on MacConkey medium that shows the selective and
differential qualities of the medium. The medium contains substances that inhibit the
growth of gram-positive bacteria. By using the lactose available in the medium, bacteria
such as Escherichia coli, Enterobacter spp., and Klebsiella spp. produce acid when they grow
on the medium, which lowers the pH of the agar and results in the formation of red to
pink colonies.

FIGURE 3-5  System for isolation of anaerobic and microaerophilic bacteria. The
packets are opened, and the sachets inside are placed into the container with the inocu-
A B
lated plates. The container is sealed immediately. The sachets contain ascorbic acid.
FIGURE 3-4  Blood culture systems. A, BACTEC system showing resin beads that bind ­Atmospheric oxygen in the container is rapidly absorbed and replaced with carbon dioxide
antimicrobial drugs. B, Pediatric, aerobic, and anaerobic BACTEC systems for blood culture in an ­exothermic reaction process. After an appropriate incubation period, the container is
together with adult and pediatric WAMPOLE Isolator tubes (front) for lysis-centrifugation opened and the plates inspected for growth. An anaerobic indicator can be placed within
culture of blood. the box to ensure that the proper conditions are maintained.
CHAPTER 3  Isolation and Identification of Aerobic and Anaerobic Bacteria 21

anaerobic culture of blood, but anaerobic bacteremia may be allowing the surface to dry, and use of a 3- to 4-mm punch
occur with other underlying causes of bloodstream infections.6 and sterile surgical instruments to collect the biopsy, which is
Common blood culture contaminants include coagulase-negative then submitted in transport medium or a small amount of sterile
staphylococci, Bacillus spp., corynebacteria, and propionibac- saline. The biopsy site can then be sutured closed.
teria. When only one blood culture is positive for one of these For deep pyoderma, a biopsy of the lesion should also be
organisms, contamination is likely, but the presence of mul- obtained after surface disinfection with 70% alcohol. The der-
tiple positive blood cultures from separate sites suggests true mis should be excised with a sterile blade and the biopsy sub-
bacteremia. mitted as just described.
Culture of intravascular catheter tips is performed to deter- For dogs and cats with wounds, aspirates and deep tissue
mine whether a catheter is a source of bacteremia and should biopsies after surgical dissection are always preferable to swab
only be performed simultaneously with blood culture. The cath- specimens. Swab specimens are often contaminated by surface
eter is removed after cleaning the skin around the catheter site microorganisms, contain only a small amount of material, and
with alcohol, and the distal 5 cm is aseptically removed into a are not amenable to optimal anaerobic transport. For this rea-
sterile tube and submitted immediately.7 In the laboratory, the son, collection of pus from fistulous tracts using a swab should
catheter tip is rolled onto an agar plate. The result of catheter also be avoided. If an abscess is present, an aspirate of the
culture alone should not change treatment. Therefore, in most abscess fluid should be collected and submitted for aerobic and
situations, blood culture may be all that is required. anaerobic culture. For dogs and cats with cellulitis, an aspirate
can be attempted by injecting a small volume of sterile saline
Skin into the lesion and aspirating back into a syringe, although
For dogs with superficial pyoderma, specimens for culture bacterial yields may be low and biopsy may be required in
should ideally be collected from a pustule. No surface disinfec- some cases.
tion should be performed. The hair should be clipped using ster-
ile scissors; clippers should be avoided. A pustule is lanced with Body Fluids
a sterile narrow-gauge needle, and pus on the needle or exuding Usually, 1 to 5 mL of body fluid is required for culture. Fluid
from the pustule is applied to a sterile swab, which can then be can be inoculated into blood culture bottles in the laboratory or
placed in transport medium for transport to the laboratory. If a alongside the patient. Body fluids that have the potential to clot
pustule cannot be found, cultures can also be obtained by lift- should be submitted to the laboratory in a tube that contains
ing crusts and swabbing beneath the crust, or rolling the swab an appropriate ratio of anticoagulant. In the laboratory, body
across an epidermal collarette two to three times. Papules can be fluids with volumes that exceed 1 mL are generally concentrated
cultured by biopsy after surface disinfection with 70% alcohol, using centrifugation and resuspension of the pellet in a smaller
volume (e.g., 0.5 mL) of supernatant before they are inoculated
onto agar.
For cerebrospinal fluid, the chance of organism isolation also
BOX 3-2 increases when the volume submitted is maximized. If possible,
at least 0.5 mL should be submitted for culture. Cerebrospinal
Suggested Procedure for Obtaining Blood Cultures fluid specimens should be immediately transported to the labo-
ratory, preferably at room temperature.
Shave site.
Clean skin with 10% povidone iodine or 1-2% tincture of Tissue Specimens
iodine, swabbing concentrically from the center; allow Attempts should be made to obtain as large a tissue specimen as
to dry (wait 1 min). possible for culture. The use of swabs to collect specimens for cul-
If a catheter must be used for specimen collection, clean ture during surgical procedures is discouraged because of insuf-
access port with 70% alcohol and allow to dry. Avoid ficient specimen size. Small pieces of tissue can be submitted in
collection from a catheter if possible. a small amount of sterile, nonbacteriostatic saline. In the labora-
Clean stopper on top of the culture tube or bottle with tory, the tissue is minced using sterile procedures to release organ-
70% alcohol; allow to dry (wait 1 min). isms before smears are prepared and media inoculated. Because
Perform venipuncture using sterile gloves to palpate the vein. an anaerobic environment can be maintained within pieces of tis-
Inoculate blood culture bottle without changing the needle. sue greater than or equal to 1 cm3 in volume, these are suitable
Space cultures based on severity of illness, before starting for anaerobic culture without oxygen-free transport. Such tissue
antimicrobial therapy. specimens may be stored at room temperature for up to 24 hours.
• Acute febrile illness: antimicrobials need to be
changed or started immediately: two sets from Airway Specimens
separate sites, all within 10 minutes. Consideration Specimens from the upper respiratory tract are generally con-
could be given to drawing a third set using a resin taminated with commensal bacteria. Aerobic bacterial culture
bottle just before administering the second dose of of a nasal wash specimen may be indicated in cats with chronic
antimicrobials (i.e., when trough drug concentra- rhinosinusitis in which an antimicrobial resistant organism is
tions are present) suspected, in particular Pseudomonas aeruginosa. Isolation of
• Acute endocarditis: 3 sets from 3 separate sites, Bordetella bronchiseptica may also have clinical significance
within 1-2 hours in dogs and cats with nasal discharge. Growth of Mycoplasma
• Nonacute disease: 3 sets from separate sites within spp. from the nasal cavity has uncertain clinical significance,
24 hours, spaced at intervals of at least 3 hours because Mycoplasma spp. can be isolated from the nasal cavities
of both healthy cats and cats with rhinosinusitis.8-11
22 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

Specimens from the lower respiratory tract may be collected


using transtracheal, endotracheal, or bronchoalveolar lavage.
Bronchial brush specimens may also be submitted for culture,
after placing them in 1 mL of sterile nonbacteriostatic saline.
The presence of intracellular bacteria on cytologic examination
is consistent with a bacterial infection, rather than contamina-
tion. Anaerobic culture could be considered for animals with
aspiration pneumonia or migrating plant awn foreign body
pneumonia.

Feces
Whole stools or swabs that contain fecal matter should be pro-
cessed within 1 to 2 hours or refrigerated as soon as possible
after specimen collection and submitted within 24 hours. The
specimens should not be contaminated with barium or urine. FIGURE 3-6  Media paddle available for screening urine for the presence of an aerobic
Rectal swabs that contain fecal material can be immersed in bacterial infection. The paddle is impregnated on each side with selective and nonselec-
transport medium until processing. Testing of more than one tive media (in this case eosin methylene blue medium, shown, and cysteine lactulose
sequential specimen may improve detection of fecal pathogens electrolyte-deficient media, on underside, respectively). The paddle is flooded with urine,
due to the low sensitivity of fecal culture methods. Selective incubated, and inspected for growth. Eosin methylene blue medium is red and selects for
media are required for isolation of Salmonella spp. Because growth of gram-negative enteric organisms. Cysteine lactose electrolyte-deficient medium
Clostridium perfringens and C. difficile are commonly detected is initially green but turns yellow in the presence of lactose-fermenting organisms.
in normal stool specimens, toxin antigen testing may be more
appropriate than culture to determine the role these organ- discarded first to clear the catheter. Urine culture should never
isms may be playing in diarrhea (see Chapter 48). Special be performed on collection bag specimens.
medium is also required for isolation of Campylobacter (see
Chapter 47). Fastidious and Unculturable Organisms
Some bacteria, such as Bartonella spp., Borrelia burgdorferi,
Urine and Leptospira spp., require special culture conditions (see
Urine should be collected using cystocentesis whenever possi- Chapters 50 to 52). Others, such as hemotropic Mycoplasma
ble. Collection of urine via sterile catheterization in male dogs species, are unculturable, and DNA amplification methods such
is acceptable when cystocentesis is not possible. Quantitative as PCR are required for detection of these organisms. Cell cul-
cultures should always be performed. For specimens obtained ture is required to propagate certain bacteria. Examples include
by cystocentesis, any level of bacterial growth is significant, Chlamydia felis and tick-borne pathogens such as Anaplasma
although most urinary tract infections contain greater than phagocytophilum.
or equal to 103 colony-forming units (CFU)/mL of bacteria.
For urine specimens obtained via catheter from male dogs, Bacterial Identification
bacterial growth of greater than or equal to 105 CFU/mL has Following aerobic or anaerobic culture, presumptive bacterial
been considered significant.12 Only aerobic bacterial culture is identification can be made on the basis of colony morphol-
routinely indicated. Anaerobic urine culture may be indicated ogy and microscopic morphology (e.g., Gram stain character-
when there is radiologic evidence of emphysematous cystitis. istics, rods or cocci). Further differentiation is generally based
Urine specimens should be refrigerated and submitted to the on a series of additional phenotypic tests, which are selected
laboratory as soon as possible. Special urine transportation based on microscopic morphology and whether the organism
tubes can be used to preserve bacteria within the specimen is gram-negative, gram-positive, aerobic, or anaerobic. Micro-
provided they are appropriate for the volume of urine col- biology laboratories use published methods to identify bacte-
lected (e.g., BD Vacutainer urine culture and sensitivity tube). ria.13 Preliminary tests used to identify gram-positive aerobes
Urine “paddles” are also available for in-house veterinary use. include the presence or absence of hemolysis on a blood agar
These allow inexpensive screening for the presence of absence plate (Figure 3-7), a positive or negative catalase test, and the
of infection (­Figure 3-6). The paddles have media impregnated production of coagulase. Identification of gram-negative aer-
on them, are designed to be incubated in the clinic, and can obes relies on whether or not the organism ferments lactose,
provide some information on the infecting bacterial species indole production, oxidase production, and the production of a
present and the number of CFU/mL. Positive paddles can then variety of other enzymes. Assessment of motility and the ability
be submitted to the laboratory for culture and susceptibility to ferment carbohydrates are also used to differentiate bacteria.
testing, although this does not always permit accurate iden- Biochemical test strips are available that allow multiple tests
tification of all bacteria present in mixed infections (see also to be performed simultaneously (e.g., API Strips, bioMérieux)
Chapter 89). (Figure 3-8), so that identification can be made within 24 to
For animals with a urinary catheter in place and a suspected 48 hours. Automated identification systems that consist of a
urinary tract infection, culture of the catheter tip is not recom- 96-well microtiter tray that contains a variety of biochemical
mended, because it is generally contaminated with commensal reagents (e.g., Sensititre, TREK Diagnostics, Cleveland) are also
organisms from the urethra. Instead, a urine specimen should in use in some veterinary diagnostic laboratories.
be obtained by aspiration from a new urinary catheter, or, if Molecular methods such as 16S rRNA gene PCR and sequenc-
feasible, using cystocentesis. If a catheter is used to collect the ing are now being used to identify organisms to the species level
specimen, several milliliters of urine should be removed and (see Chapter 5). Speciation of some mycobacteria requires analysis
CHAPTER 3  Isolation and Identification of Aerobic and Anaerobic Bacteria 23

animal’s clinical signs. Contamination is the most common


cause of false positive cultures. Isolation of only one or two
colonies of coagulase-negative staphylococci, Bacillus spp.,
Corynebacterium spp., or propionibacteria suggests contami-
nation. Isolation of large numbers of a single type of bacteria
from a normally sterile site is usually clinically significant, espe-
cially when supported by cytologic evidence of bacteria within
phagocytes.

Antimicrobial Susceptibility Testing


Importance of Antimicrobial Susceptibility Testing
Given the increasing recognition of antimicrobial resistance
among bacteria that are isolated from small animal patients,
determination of antimicrobial susceptibility can be as or more
important to the clinician than the identity of the infecting
agent. The result of antimicrobial susceptibility testing is gen-
FIGURE 3-7  Mixture of beta-hemolytic and nonhemolytic enteric bacteria growing erally available 2 to 3 days after submission of a specimen for
on blood agar. Beta-hemolysis is characterized by complete clearing of the agar around culture. In animals that are critically ill, empiric antimicrobial
each colony. therapy may already have been initiated by the time this result
is available. The susceptibility results may show that the organ-
of mycolic acid content using high-performance liquid chromatog- ism is resistant to a drug being used, in which case the drug
raphy. Recently, the availability of matrix-assisted laser desorp- should be changed to one to which the organism is susceptible.
tion ionization time-of-flight mass spectrometry instrumentation The susceptibility pattern can help to identify an alternate drug
(i.e. the Bruker Biotyper and VITEK MS systems) has revolution- when the patient does not tolerate the initial drug prescribed.
ized bacterial identification in the clinical microbiology labora- Susceptibility testing may indicate that the organism is suscep-
tory. These systems use mass spectrometry to rapidly (on average tible to a more narrow-spectrum (and generally less expensive)
in 6 minutes) and accurately (often >95%) identify organisms to antimicrobial drug than the drug initially prescribed, in which
the species level based on their chemical composition, provided a case the spectrum should be narrowed to minimize the develop-
reference spectrum for the organism is available in the database ment of antimicrobial resistance.
used for comparison. Although the instrumentation is expensive,
the ultimate cost is lower than with conventional biochemical Methods of Susceptibility Testing
methods, because of reduction in labor and reagent costs.14 Susceptibility testing can be performed using dilution methods
or diffusion methods. The minimum inhibitory concentration
Interpretation of Bacterial Culture Results (MIC) is the lowest concentration of antimicrobial drug that
inhibits visible growth of an organism over a defined incubation
Specimens That Test Negative period, most commonly 18 to 24 hours. The MIC is determined
Negative results (or lack of growth) using bacterial culture do using dilution methods, which involve exposing the organism to
not imply the absence of bacteria. Negative test results can occur twofold dilutions of an antimicrobial drug. The concentration
as a result of inadequate specimen size (such as when swabs or range used varies with the drug and the organism being tested.
aspirates are submitted), when there are inadequate organism In vitro factors that influence MIC include the composition of
numbers at the site of specimen collection, when overgrowth with the medium, the size of the inoculum, incubation conditions,
commensal organisms occurs, or when there is loss of organism and the presence of resistant bacterial subpopulations within
viability during transportation to the laboratory. The last is par- the inoculum. Standard protocols are published by the Clinical
ticularly problematic for anaerobic bacteria. In some situations, and Laboratory Standards Institute (CLSI) that specify medium
multiple bacterial species are present within a specimen, and one composition and pH, inoculum size, inoculation procedures,
species (such as Proteus spp.) swarms the plate and obscures the agar depth, and incubation conditions, as well as quality control
presence of another. The identification of multiple bacterial types requirements.13 Failure to comply with these protocols can lead
on a Gram-stained smear allows the laboratory to plan culture to erroneous results, so laboratories that adhere to CLSI stan-
methods that maximize detection of all species present. dards should be used whenever possible. MIC testing does not
Negative test results can also occur when the organism is take into account in  vivo factors such as postantibiotic effect
not inoculated onto the correct media or sufficient time is not and the effects of protein binding on antimicrobial drug activity
allowed for growth. Clinicians must consider whether fastidi- (see Chapter 6).
ous or slow-growing organisms might be present and inform
the laboratory if they are suspected. Poor laboratory qual- Dilution Methods
ity control procedures may also lead to false-negative test
results. Use of an accredited laboratory can help minimize such Dilution methods include broth macrodilution, broth micro­
problems. dilution, and agar dilution. The most widely used method
in North America is broth microdilution, whereby twofold
Specimens That Test Positive dilutions of antimicrobials are made in a broth medium
The detection of bacterial organisms within a specimen using in a microtiter plate (Figure 3-9). Pre-prepared frozen or
culture does not imply that the organism is the cause of an freeze-dried microtiter plates for inoculation are available
24 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

FIGURE 3-8  API bacterial identification strips. Only the left half of the reaction strip is shown. Top (api20STREP): Enterococcus faecium. Bottom (api20E): Escherichia coli. Positive reactions
are assigned a numerical code on the handwritten strip. The codes in each three-well group are added to generate a new (seven-number) code that can be used to identify the organism.

commercially (e.g., Sensititre plates, TREK Diagnostic Sys- interest. Agar dilution can be used to perform susceptibility
tems, Cleveland). The results can be determined using visual testing of fastidious bacteria with special medium require-
examination of the plates for the inhibition of bacterial ments, such as Campylobacter spp.
growth, or by the use of semiautomated or automated instru- The MIC for each antimicrobial drug tested against the
mentation. Dilutions can also be performed in agar, each dilu- organism is reported to the clinician on the susceptibility panel.
tion being poured onto a plate in a standardized fashion and It is the lowest concentration of antibiotic (usually in µg/mL)
allowed to set before inoculating it with the organism(s) of that inhibits growth of the organism in  vitro. The lower the
CHAPTER 3  Isolation and Identification of Aerobic and Anaerobic Bacteria 25

1 2 3 4 5 6 7 8 9 10 11 12
A AMP AMP AMP AMP AMP AMP AMP OXA+ OXA+ OXA+ OXA+ OXA+
0.25 0.5 1 2 4 8 16 0.25 0.5 1 2 4
B AUG2 AUG2 AUG2 AUG2 AMI AMI AMI AMI FOX FOX FOX FOX
4/2 8/4 16/8 32/16 4 8 16 32 2 4 8 16
C TIC TIC TIC TIC POD POD POD POD TIM2 TIM2 TIM2 TIM2
8 16 32 64 2 4 8 16 8/2 16/2 32/2 64/2
D SXT SXT SXT FOV FOV FOV FOV FOV FOV FAZ FAZ FAZ
0.5/9.5 1/19 2/38 0.25 0.5 1 2 4 8 4 8 16
E GEN GEN GEN GEN IMI IMI IMI IMI CLI CLI CLI CLI
1 2 4 8 1 2 4 8 0.5 1 2 4
F PEN PEN PEN PEN PEN PEN PEN PEN DOX DOX DOX POS
0.06 0.12 0.25 0.5 1 2 4 8 2 4 8
G XNL XNL XNL XNL XNL MAR MAR MAR MAR RIF RIF POS
0.25 0.5 1 2 4 0.25 0.5 1 2 1 2
H ENRO ENRO ENRO ENRO ERY ERY ERY ERY CHL CHL CHL POS
0.25 0.5 1 2 0.5 1 2 4 4 8 16

AMP, ampicillin; AUG2, amoxicillin/clavulanic acid (2:1 ratio); TIC, ticarcillin; SXT,
trimethoprim/sulfamethoxazole; GEN, gentamicin; PEN, penicillin; XNL, ceftiofur; ENRO, enrofloxacin;
FOV, cefovecin; AMI, amikacin; POD, cefpodoxime; IMI, imipenem; ERY, erythromycin; MAR,
marbofloxacin; OXA+, oxacillin + 2% NaCl; FOX, cefoxitin; TIM2, ticarcillin/clavulanic acid (constant 2);
CLI, clindamycin; DOX, doxycycline; CHL, chloramphenicol; FAZ, cefazolin; RIF, rifampin; POS, positive
control.
FIGURE 3-9  Broth microdilution for determination of MICs. A series of wells contain an antimicrobial drug at twofold dilutions in a standard format. One custom plate format is
shown (TREK Diagnostic Systems Sensititre Companion Animal MIC plate). A pellet of bacteria (in this case, Escherichia coli from an infected feline esophagostomy tube site) settles on the
bottom of each well when growth fails to be inhibited by the antimicrobial drug concentration tested in that well. The cutoff MICs that define susceptibility versus resistance are published
by the Clinical and Laboratory Standards Institute (CLSI) in the United States. This isolate was reported as resistant to ampicillin (>16 mg/mL), susceptible to amoxicillin-clavulanic acid
(8 mg/mL), susceptible to amikacin (8 mg/mL), susceptible to cefoxitin (8 mg/mL), resistant to ticarcillin (>64 mg/mL), susceptible to cefpodoxime (≤2 mg/mL), susceptible to ticarcillin-
clavulanic acid (16 mg/mL), resistant to trimethoprim-sulfamethoxazole (>2 mg/mL), susceptible to cefovecin (1 mg/mL), susceptible to cefazolin (≤4 mg/mL), susceptible to gentamicin
(2 mg/mL), susceptible to imipenem (≤1 mg/mL), resistant to doxycycline (>8 mg/mL), resistant to enrofloxacin and marbofloxacin (both >2 mg/mL), and resistant to chloramphenicol
(>16 mg/mL). The MIC for ceftiofur was ≤0.25 mg/mL, but because a breakpoint for this drug has not been published, only the MIC was reported, and not whether the isolate was sus-
ceptible or resistant. Results for some drugs tested, such as oxacillin, are not reported for gram-negative bacteria because these drugs are used to treat gram-positive bacterial infections.
26 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

MIC, the more potent is the antimicrobial at inhibiting bacterial combination, the organism is defined as susceptible. The break­
growth. point is not reported to the clinician. Breakpoints are established
on the basis of multiple factors (Box 3-3), which include (1) a
Diffusion Methods knowledge of MIC distributions and resistance mechanisms for
each organism-drug combination (Figure 3-11),16 (2) clinical
Diffusion methods include gradient diffusion (also known as response rates in humans and animal models, (3) how the drug
Etest) and disc diffusion. The Etest involves use of a plastic strip is distributed and metabolized in the body (pharmacokinetics),
coated with an antimicrobial gradient on one side and an MIC and (4) whether the drug is concentration dependent or time
interpretive scale on the other side (Figure 3-10, A). An agar dependent as it relates to antibacterial effect (pharmacodynam-
plate is inoculated with the organism of interest so that sub- ics) (see Box 3-3).15 Zone diameter breakpoints for disc ­diffusion
sequent growth of the organism forms a “lawn,” rather than testing are determined by correlation with MIC values. For
individual colonies (i.e., inoculation to confluence). At the time simplicity, breakpoints are established for antimicrobial drug
of inoculation, the strips are applied to the surface of the plate.
The antimicrobial drug diffuses into the medium, resulting in an
elliptical zone of growth inhibition around the strip. The MIC is
read at the point of intersection of the ellipse with the MIC scale
on the strip. Although the strips are expensive, Etests have the
advantage of being adaptable to use with fastidious organisms
and anaerobes.
Disk diffusion, also known as Kirby-Bauer antibiotic testing,
involves application of commercially available drug-impreg-
nated filter paper discs to the surface of an agar plate that has
been inoculated to confluence with the organism of interest.
Commercially available, mechanical disk-dispensing devices can
be used to apply several disks simultaneously to the surface of
the agar. The drug diffuses radially through the agar, the con-
centration of the drug decreasing logarithmically as the distance
from the disk increases. This results in a circular zone of growth
inhibition around the disk, the diameter of which is inversely
proportional to the MIC (see Figure 3-10, B). The zone diam-
eters are interpreted on the basis of guidelines ­published by A
CLSI (see Definition of Breakpoints, next), and the organisms
are reported as susceptible, intermediate, or resistant. Disk dif-
fusion can only be used to test rapidly growing organisms that
have consistent growth rates, for which criteria for interpreta-
tion of zone sizes are available.

Definition of Breakpoints
Once susceptibility testing has been performed, organisms are
classified on the susceptibility panel report as “susceptible” (S),
“resistant” (R), and, in some cases, of “intermediate” (I) sus-
ceptibility. This refers to a predicted in  vivo situation, rather
than in vitro susceptibility. The growth of “susceptible” isolates
is predicted to be inhibited by antimicrobial drug concentra-
tions that are usually achievable in blood and tissues using
normal dosage regimens. “Intermediate” isolates have MICs
that approach usually attainable blood and tissue levels and
for which response rates may be lower than those of suscep-
tible isolates. This category implies clinical efficacy in body sites
where the drugs are normally concentrated (e.g., enrofloxacin
and amoxicillin in urine) or when a higher-than-normal dose of
a drug can be used. “Resistant” isolates are predicted to grow
in the face of the usually achievable concentrations of the drug B
in blood and tissues.15
So why can the laboratory predict in vivo susceptibility on FIGURE 3-10  Alternatives to broth microdilution for antimicrobial suscepti­
the basis of MIC determination, which is an in  vitro test? In bility ­testing. A, Etest that shows susceptibility of a Bacteroides fragilis isolate to
­amoxicillin-clavulanic acid. The strip in the center is impregnated with a gradient of
order to do this, the laboratory refers to breakpoints, or clinical
­antimicrobial drug (in this case, amoxicill-clavulanic acid), with the highest concen-
­cutoff MICs (or, for disc diffusion testing, cutoff zone diam- tration at the top of the strip. The MIC is read at the point where bacterial growth is no
eters), which are established, published, and revised regularly longer inhibited by the strip (0.19 mg/mL in this case). B, Disc diffusion (also known as
by committees that are affiliated with standards agencies such as Kirby-Bauer testing) for Escherichia coli showing varying degrees of resistance to different
the CLSI. If the MIC determined in the microbiology laboratory antimicrobial drugs, reflected by zones of growth inhibition around discs containing each
is lower than a published breakpoint for that organism-drug antimicrobial drug.
CHAPTER 3  Isolation and Identification of Aerobic and Anaerobic Bacteria 27

concentrations in the bloodstream, and are based on a ­specific as “susceptible.” Urine MIC panels have been controversial
dosage regime for the antimicrobial drug tested. This dosage because the possibility of concurrent pyelonephritis (tissue drug
regime is selected by the standards agency involved. Because concentrations rather than urine concentrations) cannot always
some antimicrobials are concentrated extensively in urine, some be ruled out. Breakpoints may be reevaluated when new mecha-
veterinary laboratories may offer urine MIC ­panels, which nisms of resistance appear in bacteria or when new data are
provide breakpoints for lower urinary tract infections. These generated that improve understanding of the pharmacokinetics
breakpoints are higher than corresponding serum MIC break- and pharmacodynamics of an antimicrobial drug.
points, so a greater proportion of organisms tested are classified For infections in sites such as the central nervous system, the
clinician needs to consider whether or not (1) an antimicrobial
will penetrate that site, (2) the drug is appropriate and safe, and
(3) a higher dose is necessary to achieve adequate concentra-
BOX 3-3 tions. The clinician must also consider factors such as immuno-
suppression and other concurrent illness or drug therapy when
Factors Used by Standards Agencies to Establish treating infections on the basis of antimicrobial susceptibility
Breakpoints for Antimicrobial Susceptibility Testing test results.
The bacterial species isolated determines which antimicro-
Bacterial Factors bials the laboratory selects for inclusion in susceptibility pan-
• Evaluation of MIC distributions for each bacterial els. For example, susceptibility to cephalosporins may not be
species. These tend to be normally distributed (see reported for enterococci because of intrinsic resistance. Some
Figure 3-10; available for different antimicrobial antimicrobial drugs (e.g., vancomycin, linezolid, imipenem)
drugs at www.eucast.org).15 should be reserved for treatment of multiple-drug resistant
• Knowledge of bacterial resistance mechanisms, as organisms that cause life-threatening infections. Consultation
determined using phenotypic or genetic testing (e.g., with a health professional with expertise in infectious diseases
testing for altered penicillin binding proteins in or antimicrobial pharmacology is recommended before these
staphylococci) drugs are selected.
Pharmacokinetics of the antimicrobial drug when used at Minimum and Serum Bactericidal Concentration
a specific dosage regime in the animal species of interest,
including protein binding, taking into account variation The minimum bactericidal concentration (MBC) is the mini-
among individuals mum concentration of an antimicrobial drug that is bacteri-
Pharmacodynamic indices (e.g., whether killing is concen- cidal. It is determined by re-culturing (subculturing) broth
tration or time dependent) for the antimicrobial drug as dilutions that inhibit growth of a bacterial organism (i.e., those
they relate to organism killing (see Chapter 6) at or above the MIC). The broth dilutions are streaked onto
Clinical response rates in humans and animal models, agar and incubated for 24 to 48 hours. The MBC is the low-
which are based on prospective, randomized controlled est broth dilution of antimicrobial that prevents growth of the
clinical studies with well-defined dosage regimes organism on the agar plate. Failure of the organism to grow
on the plate implies that only nonviable organisms are present.
The use of MBCs has been advocated by some for treatment

FIGURE 3-11  Histogram that shows the distribution of amikacin minimum inhibitory concentrations (MICs) for isolates of Escherichia coli. The MICs are normally distributed. (From
www.eucast.org. Accessed May 11, 2012.)
28 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

of serious infections (such as endocarditis) or for treatment of 3. Cole LK, Kwochka KW, Kowalski JJ, et  al. Microbial flora and
immunosuppressed patients. Their value has been controver- antimicrobial susceptibility patterns of isolated pathogens from the
sial, and they are not widely performed in human or veterinary horizontal ear canal and middle ear in dogs with otitis media. J Am
medicine. Vet Med Assoc. 1998;212(4):534-538.
4. Tolar EL, Hendrix DV, Rohrback BW, et  al. Evaluation of
Serum bactericidal testing (SBT) is performed in the same
clinical characteristics and bacterial isolates in dogs with bac-
manner as the MBC test, but uses the serum of a patient that has terial keratitis: 97 cases (1993-2003). J Am Vet Med Assoc.
been treated with an antimicrobial. Serum is usually ­collected 2006;228(1):80-85.
at expected peak and trough serum concentrations. SBC can be 5. Clinical and Laboratory Standards Institute. http://www.clsi.org:
used to determine whether a patient has been adequately dosed, Last accessed May 15, 2012.
and to ensure that the antimicrobial is having the desired effect 6. Sykes JE, Kittleson MD, Pesavento PA, et al. Evaluation of the rela-
on the organism. Some data are available to support specific tionship between causative organisms and clinical characteristics of
peak and trough SBT results required for the optimal treatment infective endocarditis in dogs: 71 cases (1992-2005). J Am Vet Med
of human endocarditis, osteomyelitis, and cancer chemotherapy Assoc. 2006;228(11):1723-1734.
patients.17-20 The SBT has not been widely used or evaluated for 7. Thomson RB. Specimen collection, transport, and processing:
bacteriology. In: Murray PR, Barron EJ, Jorgensen JP, et  al. eds.
infections in dogs and cats and remains controversial in human
Manual of Clinical Microbiology. 9th ed. Washington, DC: ASM
medicine. Press; 2007:291-333.
8. Veir JK, Ruch-Gallie R, Spindel ME, et al. Prevalence of selected
Mutant Prevention Concentration infectious organisms and comparison of two anatomic sampling
The mutant prevention concentration (MPC) is the lowest anti- sites in shelter cats with upper respiratory tract disease. J Feline
microbial drug concentration required to block the growth of Med Surg. 2008;10(6):551-557.
the least susceptible bacterial cell in high density bacterial pop- 9. Johnson LR, Drazenovich NL, Foley JE. A comparison of routine
ulations.21 It is the MIC of the most resistant bacterial strain culture with polymerase chain reaction technology for the detection
in a mixed bacterial population. Concentrations of antimicro- of Mycoplasma species in feline nasal samples. J Vet Diagn Invest.
bial between the MIC and the MPC allow selective amplifica- 2004;16(4):347-351.
10. Randolph JF, Moise NS, Scarlett JM, et al. Prevalence of mycoplas-
tion of resistant mutants (also known as the mutant selection
mal and ureaplasmal recovery from tracheobronchial lavages and
window).22 This range is considered to be a “danger zone” of mycoplasmal recovery from pharyngeal specimens in cats with
for therapeutic drug concentrations. Minimizing the length of or without pulmonary disease. Am J Vet Res. 1993;54:897-900.
time that the antimicrobial drug concentration remains in the 11. Tan RJS, Lim EW, Ishak B. Ecology of mycoplasmas in clinically
mutant selection window in vivo may reduce the likelihood for healthy cats. Aust Vet J. 1977;53:515-518.
resistance selection during treatment.22 The MPC cannot be 12. Ling GV. Lower Urinary Tract Diseases of Dogs and Cats. St Louis:
extrapolated from the MIC. It is estimated using the standard Mosby; 1995:116.
agar dilution method used to estimate MIC, but with a larger 13. Murray PR, Barron EJ, Jorgensen J, et al. eds. Manual of Clinical
inoculum (≥109 instead of 105 organisms), so as to include Microbiology. 9th ed. Washington, DC: ASM Press; 2007.
resistant subpopulations of bacteria. Administration of higher 14. Bizzini A, Greub G. Matrix-assisted laser desorption ionization
time-of-flight mass spectrometry, a revolution in clinical microbial
doses of antimicrobial drugs that exceed the MPC increases
identification. Clin Microbiol Infect. 2010;16(11):1614-1619.
the chance of toxicity to the patient, but offsets the chance of 15. Turnidge JL, Paterson DL. Setting and revising antimicrobial sus-
selection for resistant organisms, even though infection may ceptibility breakpoints. Clin Microbiol Rev. 2007;20(3):391-408.
be cured with lower doses.22,23 Although first evaluated for 16. The European Committee on Antimicrobial Susceptibility Testing—
the fluoroquinolones, data is accumulating regarding the use- EUCAST. http://www.eucast.org. Last accessed May 14, 2012.
fulness of the MPC for other drug classes. MPC estimations 17. Klastersky J, Daneau D, Swings G, et  al. Antibacterial activity
are currently not routinely performed in veterinary diagnostic in serum and urine as a therapeutic guide in bacterial infections.
laboratories. J Infect Dis. 1974;129(2):187-193.
18. Reller LB. The serum bactericidal test. Rev Infect Dis. 1986;
SUGGESTED READINGS 8(5):803-808.
19. Weinstein MP, Stratton CW, Ackley A, et al. Multicenter collaborative
Drlica K, Zhao X. Mutant selection window hypothesis updated. Clin evaluation of a standardized serum bactericidal test as a prognostic
Infect Dis. 2007;44(5):681-688. indicator in infective endocarditis. Am J Med. 1985;78(2):262-269.
Turnidge JL, Paterson DL. Setting and revising antimicrobial suscepti- 20. Weinstein MP, Stratton CE, Hawley HB, et  al. Multicenter col-
bility breakpoints. Clin Microbiol Rev. 2007;20(3):391-408. laborative evaluation of a standardized serum bactericidal test as a
predictor of therapeutic efficacy in acute and chronic osteomyelitis.
REFERENCES Am J Med. 1987;83(2):218-222.
1. Radaelli ST, Platt SR. Bacterial meningoencephalomyelitis in dogs: 21. Blondeau JM, Hansen G, Metzler K, et  al. The role of PK/PD
a retrospective study of 23 cases (1990-1999). J Vet Intern Med. parameters to avoid selection and increase of resistance: mutant
2002;16(2):159-163. prevention concentration. J Chemother. 2004;16(suppl 3):1-19.
2. Sturges BK, Dickinson PJ, Kortz GD, et  al. Clinical signs, mag- 22. Drlica K, Zhao X. Mutant selection window hypothesis updated.
netic resonance imaging features, and outcome after surgical and Clin Infect Dis. 2007;44(5):681-688.
medical treatment of otogenic intracranial infection in 11 cats and 23. Zhao X, Drlica K. A unified anti-mutant dosing strategy. J Antimi-
4 dogs. J Vet Intern Med. 2006;20(3):648-656. crob Chemother. 2008;62(3):434-436.
CHAPTER 4

Isolation and Identification of Fungi


Jane E. Sykes and Shelley C. Rankin

w KEY POINTS
• F ungi often grow more slowly than bacteria and can have spe- • S pecimens should be collected and transported as for bacterial
cific culture media requirements. Some fungi are dangerous to culture. Refrigeration inhibits growth of some fungi.
culture because they produce airborne spores that may lead to • Because of increasing drug resistance among pathogenic fungi,
laboratory-acquired infections. the demand for antifungal susceptibility testing has increased,
• The clinician should tell microbiology laboratory personnel if and for some fungal pathogens, culture and susceptibility
a fungal infection is suspected. Providing a list of suspected testing before beginning antifungal drug therapy may be
pathogens facilitates safe and successful culture of pathogenic advantageous.
fungi. The animal’s clinical abnormalities, travel history, history
of immunosuppressive drug therapy, and the results of cytol-
ogy or histopathology should be taken into account.

INTRODUCTION AND DEFINITIONS can exist in more than one asexual form, and therefore have more
than two names. For example, Pseudallescheria boydii (teleomorph)
Fungi are eukaryotic microorganisms that secrete enzymes in has two asexual forms, Graphium fruticola and Scedosporium apio-
order to digest organic material in the environment. The resul- spermum. Currently, there is a move by mycologists to identify only
tant nutrients are absorbed through a specialized fungal cell wall a single name for a fungal organism (‘one fungus = one name’).
(known as chemotrophic nutrition). A basic understanding of fun- Throughout this book, the name most commonly used in the lit-
gal terminology facilitates communications between clinicians, erature is used to refer to different fungal pathogens.
microbiologists, and pathologists. Fungal organisms have two
basic morphologic forms: Specimen Collection and Transport
1. Molds—molds exist as filaments known as hyphae, which may
be divided into cellular compartments (septate hyphae). A mass Isolation of fungal organisms from clinical specimens can be
of filaments is known as a mycelium. A granule formed by a com- facilitated by the use of special media. For many fungal spe-
pacted mat of fungal filaments is a eumycetoma (as opposed to cies, this may be hazardous to laboratory personnel. Submission
an actinomycotic mycetoma; see Chapter 42). of specimens for culture of dangerous organisms such as Blas-
2. Yeasts—these are single-celled organisms that reproduce by tomyces dermatitidis, Coccidioides immitis, and Histoplasma
budding. In some cases, buds do not detach from the parent cell,
but instead form a chain. Elongated chains of buds may resemble
hyphae. These are termed pseudohyphae (Figure 4-1).
Many pathogenic fungi exist as a mold in the environment,
but in tissues convert to a yeast form. These fungi are called
dimorphic fungi. An example of a dimorphic fungus is Blastomyces
dermatitidis.
Fungi reproduce by producing spores. Spores can be produced
following asexual or sexual reproduction. Asexual reproduction
(through mitosis) generates spores that are identical to the parent
cell. Sexual reproduction involves the fusion of haploid nuclei from
two hyphal structures, which is followed by meiosis. The asexual
stage of a fungus is known as an anamorph, whereas the sexual
stage is known as a teleomorph. Many fungi have two different spe-
cies names to reflect these stages. For example, the teleomorph
of Cryptococcus neoformans is known as Filobasidiella neoformans.
Formally, the teleomorph name is supposed to take precedence
and can be used to describe both stages.1 However, asexual repro-
duction predominates in most fungi, including during growth in FIGURE 4-1  Histopathology of the thyroid gland from a 13-year old female spayed
culture. Thus, certain fungi, such as C. neoformans, are more widely poodle mix with disseminated Candida albicans infection. The dog had been treated with
known by their anamorph name. In addition, teleomorphs of some prednisone, cyclosporin, and azathioprine for refractory immune-mediated hematologic
fungal organisms have not yet been described. Some organisms disease. Note intralesional pseudohyphae. H&E stain, 400× magnification.

29
30 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

capsulatum is discouraged if the diagnosis can be obtained using and molds tend to be gram negative.4 A potassium hydroxide
other methods such as cytology, histopathology, and serologic (KOH) preparation may be performed to examine a specimen
testing. These organisms exist as yeasts in tissues (at 37°C) that for dermatophyte hyphae and arthroconidia. The KOH clears
are minimally hazardous to clinicians and laboratory person- keratinaceous and cellular debris, but fungal elements are left
nel, but grow as hyphae at lower temperatures and on culture intact. If tissue granules are present, the laboratory will crush
medium, which generate spores that can be inhaled and cause them, examine them for hyphae, and the granules can also be
disease. cultured. Fungal morphology may be altered in specimens from
Specimens that are commonly submitted for fungal culture dogs and cats receiving antifungal drug treatment (Figure 4-4).
include lower respiratory tract wash specimens, cerebrospinal Special stains used in the laboratory for fungal identification
fluid, urine, other body fluids, skin, eye, hair, or biopsy speci- include India ink, which is used to identify cryptococcal yeasts
mens from the nasal cavity, bone, or skin. Guidelines for speci- in cerebrospinal fluid (see Figure 62-8), and calcofluor white,
men collection are the same as for bacterial culture (refer to which binds polysaccharides in the fungal cell wall and fluo-
Chapter 3). The clinician should inform the laboratory what resces when examined using fluorescence microscopy ­(Figure
fungal species are suspected, because different fungi have differ- 4-5, A). Other stains, such as lactophenol cotton blue (see
ent media preferences and growth conditions. As large a speci- ­Figure 4-5, B), are also used to identify fungi after they have
men as possible should be collected. Care should be taken to been grown in culture.
avoid collection of contaminated specimens. Blood cultures may In some situations, fungi are observed in tissues with histo-
be useful for dogs and cats with disseminated fungal disease pathology but a culture of the causative organism is not avail-
when lesions cannot be easily accessed. Detection of fungemia able, either because it has not been performed or because the
can sometimes be achieved using blood culture systems for bac- organism has failed to grow. The morphology and staining
terial isolation. Lysis-centrifugation methods (Isolator tubes, characteristics of the organism on histopathology may also be
Wampole Laboratories) may be more sensitive for detection of useful for preliminary identification or allow full identification
dimorphic and filamentous fungi in blood. Mycobacterial blood of the pathogen. Special stains used include Mayer’s mucicar-
culture media (such as MB/BacT, BioMérieux) may also offer mine stain, which stains the cryptococcal polysaccharide cap-
increased sensitivity for detection of fungemia.2 sule red; Gomori’s methenamine silver stain, which stains fungal
For dermatophyte culture, 10 to 12 hairs should be plucked organisms black; and periodic acid–Schiff, which stains fungal
from the edge of the lesion using sterile forceps and submit- elements dark pink (Figure 4-6). If the identity of the fungus
ted without refrigeration in a clean dry paper envelope or ster- remains in doubt, an additional specimen should be collected
ile glass or plastic tube. Paper envelopes are preferred to tubes for culture if possible, and the microbiology laboratory should
because tubes retain moisture that can promote overgrowth of be made aware of the morphology of the organism and the dif-
contaminants. Scrapings of crusts at the edges of the lesion can ferential diagnoses. Complete identification of the organism
also be submitted. Claw clippings can be collected after cleaning using culture is useful for proper treatment and prognostication.
them with 70% alcohol to remove contaminating bacteria and
fungi. Crumbling material from under the claw may be the best Fungal Culture
specimen, or the entire claw can be submitted. Before culture, clinical specimens must be prepared for inocula-
If transport requires longer than 2 hours, appropriate trans- tion. For example, body fluids are usually first centrifuged by
port media should be used or specimens should be refrigerated. the laboratory to concentrate fungal organisms. The sediment is
When rapid transport to the laboratory is possible, storage and then resuspended in a smaller volume of supernatant and inocu-
transport at room temperature is preferred. Fungal culture of lated onto selective media. If biopsy specimens are submitted,
specimens that are more than 24 hours old is not recommended the tissues are finely sliced, ground, or minced, which releases
because of increased likelihood of false-positive and false-nega- the organisms before they are inoculated onto the media.
tive results. The laboratory should be informed if the patient is Many types of media are available for isolation of fungi,
receiving antifungal drug treatment, which may interfere with and no single medium is optimal for all organisms. Many
the ability to culture fungus from the specimen. fungi grow on blood agar plates used for primary isolation
Specimens should be labeled with the patient’s name, patient of bacteria. Special media used for initial (primary) isolation
number, and source of specimen, as well as the date and time of fungal organisms from tissues and body fluids are shown
of collection, and labeled, packaged, and transported accord- in Table 4-2. Other media are available for differentiation of
ing to regional and global regulations for shipping of infectious fungal species (see Fungal Identification, later). Media that
substances (see Chapter 1).3 Careful packaging and transport contain antibacterial agents (usually chloramphenicol or gen-
is particularly important for specimens suspected to contain tamicin) are used to culture specimens that might contain bac-
fungal pathogens. If dangerous organisms might be present, teria. Cycloheximide is added to dermatophyte test medium
the laboratory should be warned in advance and the specimens to inhibit the growth of contaminant saprophytic mold spe-
marked appropriately. cies. This allows selective isolation of dermatophytes such as
Microsporum canis.
Diagnostic Methods Plates for fungal culture are sealed with an air-permeable
tape in order to prevent exposure of laboratory personnel to
Microscopic Examination of Direct Smears potentially dangerous spores and to minimize plate contami-
Examination of a direct smear before culture allows rapid par- nation with airborne fungal organisms. Plates that grow fila-
tial or complete identification of many fungi by the laboratory mentous fungi are opened only in a biological safety cabinet.
(Table 4-1, Figure 4-2). This can help the clinician make initial Laboratories prefer to use media in tubes or bottles to reduce
treatment decisions and guides media selection by the labora- occupational exposure to aerosolized fungal elements. Fungal
tory. Using Gram stain, yeasts generally stain gram positive cultures are incubated at room temperature (22°C to 25°C)
CHAPTER 4  Isolation and Identification of Fungi 31

TABLE 4-1
Microscopic Descriptions of Some Common Fungal Pathogens of Dogs and Cats
Yeast or Geographic
Organism Hyphae Diameter (µm) Description Distribution
Blastomyces Yeast 8-15 Round with thick, often refractile cell Southeastern and south
dermatitidis wall and broad-based budding (see central United States,
Figure 60-8). upper Midwest region
Histoplasma Yeast 2-5 Oval to round, often found in clusters Ohio and Mississippi
capsulatum within macrophages (see Figure 61-7). river valleys, especially
in southern United
States. Also Central
America
Cryptococcus spp. Yeast 2-15 Round to oval, narrow-based bud- Worldwide, common in
ding, sometimes forming chains of Australia and western
organisms. Large capsule (see Figures North America
4-4 and 62-8). Poorly encapsulated
variants are rarely encountered.
Coccidioides spp. Spherule 10-200 Spherules vary in size and may contain Central California,
endospores (“pomegranate-like”). ­Arizona, Nevada,
Using cytology, they appear as empty Central and South
and collapsed, deeply basophilic America
structures (see Figure 63-8).
Sporothrix Yeast 2-6 Oval, round, or cigar-shaped, occasion- Temperate and tropi-
schenckii ally budding (see Figure 64-4). May cal zones worldwide.
be found intracellularly and extra­ Most common in
cellularly. Difficult to find in dogs South and Central
but may be abundant in cats. America
Malassezia spp. Yeasts and 3-8 (yeast), 5-10 Round to oval; when budding, have Worldwide
­pseudohyphae (pseudohyphae) the appearance of “footprints” (see
Figure 59-2).
Candida spp. Yeast and 3-4 (yeast), 5-10 Usually yeasts with a single bud Worldwide
­pseudohyphae (pseudohyphae) or pseudohyphae with multiple
­constrictions (see Figure 67-3).
Dermatophytes Hyphae 3-15 Septate hyaline hyphae, arthroconidia Worldwide
may be visible along hair shafts (see
Figure 58-8)
Aspergillus spp. Hyphae 4-6 Septate hyaline hyphae with uniform Worldwide
diameter that branch at 45° angles
(see Figure 4-2).
Scedosporium spp., Hyphae 3-12 Septate hyaline hyphae that tend to Worldwide
Paecilomyces spp. branch at wider angles than Aspergillus
spp. (45° to 90°) (see Figure 4-6).
Basidiobolus and Co- Hyphae 10-30 Wide, non- or poorly septate irregular Tropical and subtropical
nidiobolus (zygomy- hyaline hyphae, branching at right regions of the
cosis) angles, may be twisted or folded. Americas, Africa,
Poorly stained using hematoxylin southeast Asia, and
and eosin stain (see Figure 4-3). Australia
Phialophora, ­Exophiala, Hyphae 1.5-6 Pigmented hyphae, budding cells with Worldwide
­Cladophialophora, single septa and chains of swollen
Bipolaris (phaeohy- round cells (see Figure 68-5). May
phomycosis or dema- form granules.
tiaceous molds)
Pythium insidiosum Hyphae 4-9 Sparsely septate, hyaline hyphae with Tropical, subtropical,
90° branches, detectable using hema- and some temperate
toxylin and eosin stain regions of the world.
32 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

FIGURE 4-2  Direct smear of an aspirate from a mediastinal mass of a 6-­year-old FIGURE 4-3  Histopathology showing severe pulmonary zygomycosis at necropsy
female spayed labrador retriever with disseminated Aspergillus deflectus infection. Note in an 11-year-old mixed-breed dog that had been diagnosed with diabetic ketoacidosis
hyphae with parallel walls that branch at 45° angles. Stain uptake by some hyphae is poor. before euthanasia. H&E stain, 100× magnification.
Wright’s stain, 1000× oil magnification. (Image courtesy Dr. William ­Vernau, University of
California, Davis.)

A
A

B
FIGURE 4-5  Special stains used for identification of fungal isolates following direct
B
smear preparation. A, Calcofluor white stain of a mass of Aspergillus fumigatus hyphae.
FIGURE 4-4  A, Cerebrospinal fluid cytology (Cytospin preparation) from a dog with 1000× oil magnification. B, Lactophenol cotton blue preparation of the mold phase of
newly diagnosed cryptococcosis. Note the thick capsule, which appears as a halo around Histoplasma capsulatum. 1000 × oil magnification.
the organism, and narrow-based budding. Wright-Giemsa stain, 1000× oil magnification.
B, Cytology of a lymph node aspirate from a cat that had been treated for cryptococco-
sis with antifungal drugs. The architecture of the organisms is distorted. Diff Quik stain, 
1000 × oil magnification.
CHAPTER 4  Isolation and Identification of Fungi 33

A B
FIGURE 4-6  Histopathology of the kidney from a 2-year-old female spayed German shepherd with disseminated paecilomycosis. A, Periodic acid Schiff stain. The fungal hyphae stain
bright pink. B, Gomori’s methenamine silver stain, which stains the fungal hyphae black. 400× magnification.

TABLE 4-2
Special Medium Types Used to Culture Fungi That Infect Dogs and Cats
Type of Medium Additives Indications
Sabouraud dextrose agar None Cultivation of all fungi.
Brain-heart infusion agar None Cultivation of all fungi.
Potato dextrose agar or potato None Cultivation of all fungi. Especially useful for opportu-
flake agar nistic molds. Induces sporulation.
Inhibitory mold agar Chloramphenicol, sometimes gentamicin Cultivation of all fungi when bacterial contamination
is suspected or possible.
Dermatophyte test medium Chloramphenicol, gentamicin, Dermatophyte isolation. Shows a color change when
cycloheximide dermatophytes grow on the medium.

or 30°C, a lower temperature than that used for routine cul- fungi. A variety of broth or solid media are also used for fun-
ture of bacteria. In general, cultures should be examined daily gal identification. For example, Candida species can be differ-
for 2 weeks and twice weekly for an additional 2 to 4 weeks, entiated using Candida bromcresol green (BCG) agar, because
because the growth of some organisms may be very slow. In each species has a specific colonial morphology and color on
general, rapidly growing fungi grow in 1 to 3 days, those with this medium. Test strips are commercially available that allow
intermediate growth in 5 to 9 days, and slow growers take up multiple biochemical identification tests for yeasts to be per-
to 4 weeks. Yeast colonies are smooth and resemble bacterial formed simultaneously (e.g., API 20C AUX Yeast Identification
colonies, and molds grow in colonies that are dry, wrinkled, or Kit, bioMérieux) (see Figure 3-8), so identification can be made
heaped ­(Figure 4-7). within 1 to 2 days.

Fungal Identification Interpretation of Fungal Culture Results


After growth in culture, fungi are identified based on visual
characteristics such as colony morphology and color. Light False-negative and false-positive results are very common with
microscopy is useful to evaluate the microscopic morphology fungal culture, and results must be interpreted in light of the
of yeasts and to determine the presence of septate or nonseptate organism grown, the animal’s clinical condition, and the results
hyphae and fruiting structures for molds (Table 4-3). Increas- of cytology and histopathology.
ingly, DNA sequence information has been used to identify
fungi (such as after PCR of D2 or ITS regions of the 23S rRNA Specimens That Test Negative
gene). Matrix-assisted laser desorption ionization time-of-flight Negative test results can occur as a result of inadequate speci-
(MALDI-TOF) mass spectrometry holds considerable promise men size (such as when swabs or aspirates are submitted), when
for identification of fungi in the future (see Chapter 3).5 Molds there are inadequate organism numbers at the site of specimen
that produce pigment are known as dematiaceous fungi (see collection, when overgrowth with contaminating bacteria or
Figure 4-7, B), and those that do not are known as hyaline saprophytic fungi occurs, or when there is loss of organism
34 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

viability during specimen transportation. If the patient is receiv-


ing antifungal drugs but has not completely cleared the infec-
tion, culture may be negative.
False-negative test results also occur when the organism is
not inoculated onto the correct media or sufficient time is not
allowed for growth. It is critical that clinicians inform the lab-
oratory if a fungal infection is suspected, and what organism
might be involved. Poor laboratory quality control procedures
may also lead to false-negative test results. Use of an accredited
laboratory can help minimize such problems.

Specimens That Test Positive


The detection of fungal organisms within a specimen does not
always imply that the organism is causing the animal’s clinical
signs. Contamination by saprophytic fungi, especially Aspergil-
lus, is the most common cause of false-positive fungal cultures.
Organisms are more likely to be clinically significant when cyto-
A logic examination of a stained smear also demonstrates the pres-
ence of fungal organisms.

Antifungal Susceptibility Testing


Increasing resistance to antifungal agents has been docu-
mented for a variety of fungal pathogens. This has led to
an increased demand for antifungal susceptibility testing by
clinicians. Fortunately, the reproducibility of antifungal sus-
ceptibility testing has improved greatly in recent years, and
breakpoints (see Chapter 3) have been established for treat-
ment of many fungal infections in human patients. Although
breakpoints are still lacking for veterinary patients, antifun-
gal susceptibility testing allows comparison of the activity of
two or more antifungal drugs, and serial testing of isolates
B from a single patient allows the development of resistance to
FIGURE 4-7  A, Growth of Cryptococcus gattii yeasts on potato flake agar. B, Clado- be monitored over time. Methods of susceptibility testing that
phialophora bantiana, a pigmented (dematiaceous) mold, isolated from canine brain tis- have been adapted for yeast and mold infections include broth
sue on potato flake agar. microdilution, the Etest, and disk diffusion (see Chapter 3).

TABLE 4-3
Classification of Fungi That Are Known to Infect Dogs and Cats
Phylum Class Example Characteristics
Zygomycota Zygomycetes Absidia, Rhizopus, Conidiobolus, Aseptate hyphae. In asexual reproduction, spores
Basidiobolus are abundantly produced in a closed sac called
a sporangium. Sexual reproduction results in a
single, large thick-walled zygospore.
Basidiomycota Basidiomycetes Cryptococcus, Malassezia Sexual reproduction leads to haploid production
of basidiospores on a basidium.
Ascomycota Archiascomycetes Pneumocystis Sexual reproduction leads to haploid production
Hemiascomycetes Candida spp. of ascospores in a sac-like structure (ascus).
Euascomycetes Histoplasma, Blastomyces, Organisms are more commonly identified based
­Microsporum, Trichophyton on characteristics of asexual reproduction.
Aspergillus spp.
Fusarium spp.
Pseudallescheria spp. (teleomorph
of Scedosporium spp.)
CHAPTER 4  Isolation and Identification of Fungi 35

Common antifungal agents tested include amphotericin B, a wide range of antifungal drug dilutions may also occur
itraconazole, fluconazole, voriconazole, posaconazole, caspo- (a phenomenon known as trailing). Criteria have been set for
fungin, griseofulvin, and flucytosine (Figure 4-8). Limitations interpretation of MICs in the presence of trailing for differ-
of antifungal susceptibility testing include the slow growth ent antifungal drugs.6 MALDI-TOF mass spectrometry can be
of fungal pathogens when compared to bacteria, and the fact used to determine antifungal drug susceptibility among fungal
that fungi may grow as yeasts or molds depending on media pathogens and in future may overcome some of the difficulties
and incubation conditions. Partial inhibition of growth over that currently exist.7

1 2 3 4 5 6 7 8 9 10 11 12
A POS AND AND AND AND AND AND AND AND AND AND AB
0.015 0.03 0.06 0.12 0.25 0.5 1 2 4 8 0.12
B MF MF MF MF MF MF MF MF MF MF MF AB
0.008 0.015 0.03 0.06 0.12 0.25 0.5 1 2 4 8 0.25
C CAS CAS CAS CAS CAS CAS CAS CAS CAS CAS CAS AB
0.008 0.015 0.03 0.06 0.12 0.25 0.5 1 2 4 8 0.5
D FC FC FC FC FC FC FC FC FC FC FC AB
0.06 0.12 0.25 0.5 1 2 4 8 16 32 64 1
E PZ PZ PZ PZ PZ PZ PZ PZ PZ PZ PZ AB
0.008 0.015 0.03 0.06 0.12 0.25 0.5 1 2 4 8 2
F VOR VOR VOR VOR VOR VOR VOR VOR VOR VOR VOR AB
0.008 0.015 0.03 0.06 0.12 0.25 0.5 1 2 4 8 4
G IZ IZ IZ IZ IZ IZ IZ IZ IZ IZ IZ AB
0.015 0.03 0.06 0.12 0.25 0.5 1 2 4 8 16 8
H FZ FZ FZ FZ FZ FZ FZ FZ FZ FZ FZ FZ
0.12 0.25 0.5 1 2 4 8 16 32 64 128 256

POS, positive control; AND, anidulafungin; MF, micafungin; CAS, caspofungin; FC, 5-flucytosine; PZ,
posaconazole; VOR, voriconazole; IZ, itraconazole; FZ, fluconazole; AB, amphotericin B.
B
FIGURE 4-8  A, Minimum inhibitory concentration (MIC) assay for a Candida krusei isolate. Growth in the presence of the antifungal drug is characterized by formation of a pellet on
the bottom of the well and a change in the color of the medium from purple to pink. B, Schematic of the same microtiter plate showing antifungal drug dilutions. Organisms are defined
as susceptible, intermediate, or resistant based on MIC cutoffs (breakpoints or interpretive criteria) set by regulatory bodies such as the Clinical and Laboratory Standards Institute in the
United States. The report to the clinician would read susceptible to anidulafungin (0.12 µg/mL), susceptible to micafungin (0.25 µg/mL), susceptible to caspofungin (1 µg/mL), intermediate
susceptibility to flucytosine (8 µg/mL), susceptible to voriconazole (0.12 µg/mL), susceptible to itraconazole (0.25 µg/mL), and resistant to fluconazole (128 µg/mL). The MICs for posacon-
azole and amphotericin B were reported as 0.25 µg/mL and ≤0.12 µg/mL, but the organism was not classified as susceptible or resistant because interpretive criteria for this organism-drug
combination were not available.
36 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

REFERENCES 5. Iriart X, Lavergne RA, Fillaux J, et  al. Routine identifica-


tion of medical fungi by MALDI-TOF: performance of the new
1. Pitt JI, Samson RA. Nomenclatural considerations in naming spe- VITEK MS using a new time-effective strategy. J Clin Microbiol.
cies of Aspergillus and its teleomorphs. Stud Mycol. 2007;59:67-70. 2012;50(6):2107-2110.
2. Taniguchi T, Ogawa Y, Kasai D, et al. Three cases of fungemia in 6. Johnson EM, Espinel-Ingroff AV, Pfaller MA. Susceptibility test
HIV-infected patients diagnosed through the use of mycobacterial methods: yeasts and filamentous fungi. In: Versalovic J, Carroll
blood culture bottles. Intern Med. 2010;49:2179-2183. KC, Funke G, et al. eds. Manual of Clinical Microbiology. 10th ed.
3. World Health Organization. A guide for shipping infectious Washington, DC: ASM Press; 2011:2020-2037.
­substances. 2009. http://www.who.int/ihr/infectious_substances/en/ 7. Marinach C, Alanio A, Palous M, et  al. MALDI-TOF MS-based
index. Last accessed May 11, 2012. drug susceptibility testing of pathogens: the example of Candida
4. Shea YR. General approaches for direct detection of fungi. In: Ver- albicans and fluconazole. Proteomics. 2009;9:4627-4631.
salovic J, Carroll KC, Funke G, et al. eds. Manual of Clinical Micro-
biology. 10th ed. Washington, DC: ASM Press; 2011:1776-1792.
CHAPTER 5

Nucleic Acid Detection Assays


Jane E. Sykes and Shelley C. Rankin

w KEY POINTS
• A  ssays for detection of nucleic acid such as polymerase chain • I nterpretation of the results of molecular diagnostic tests
reaction assays are increasingly available commercially for requires knowledge of the problems and pitfalls of each type
diagnosis of companion animal infectious diseases. of assay. The results of testing at one laboratory may differ from
• Molecular diagnostic assays include those that involve amplifi- that at another because of variations in assay design.
cation (the copying of nucleic acid so that it can be more read- • With increased automation, laboratory proficiency testing,
ily detected) or probe-based assays that do not involve nucleic improved quality assurance, and standardization of reporting,
acid amplification. the availability and utility of molecular diagnostic tests for com-
• Specimens for these assays should be collected using sterile panion animal infectious diseases will increase.
technique in order to minimize contamination.

INTRODUCTION A plethora of nucleic acid detection methods have been devel-


oped since the advent of PCR, many of which are commercially
Assays that detect nucleic acid, also known as molecular diagnos- available for diagnosis of human infectious diseases but are not
tic assays, include probe-based assays such as in situ hybridiza- yet being widely used in veterinary medicine. These are described
tion, and methods that amplify DNA, such as the polymerase chain in detail elsewhere.1,3,4 The purpose of this chapter is to outline
reaction (PCR). (1) the indications for nucleic acid detection methods, (2) the
DNA or RNA probes are short, single-stranded nucleic acids best ways to collect and transport specimens for these assays, (3)
(usually <50 base pairs) that have a sequence that hybridizes to the most commonly used methods in veterinary medicine, and
(i.e., is complementary to) a specific segment of DNA from the tar- (4) guidelines for their interpretation.
get microorganism (Figure 5-1). For in situ hybridization, the probe
is labeled with a fluorescent or chemical tag so that the bound Specimen Collection and Transport
probe can be detected. The probe is reacted with a tissue specimen
on a microscope slide in order to determine if the organism is pres- The recommendations for specimen collection and transport pre-
ent in the specimen. For PCR, two specific short (approximately 20 sented here are based on the guidelines published by the Clinical
bases) DNA sequences called primers, which are complementary and Laboratory Standards Institute (CLSI).5 These guidelines are
to the target microorganism’s DNA, are used in conjunction with updated on a regular basis and ensure that detection of nucleic
a machine called a thermocycler to amplify a specific segment of acids within clinical specimens is optimal. In some instances,
DNA from just one copy to millions of copies that can be more eas- nucleic acid may be present and detectable in a specimen even
ily detected. In situ PCR is a combination of these two techniques.1 when the specimen has not been handled in a manner that is con-
Nucleic acid detection methods are ideally suited to detection sistent with the guidelines. Many laboratories publish their own
of organisms that are not easily found using cytology or histo- specific guidelines for specimen collection and transport. Speci-
pathology, are slow-growing, or are difficult to culture, as well as mens should be properly packaged and labeled (see Chapter 3).
when a rapid (<12 hours) diagnosis is required. Other applications
are shown in Box 5-1. Nucleic Acid Probe Assays
The use of nucleic acid–based assays for diagnosis has In situ hybridization is generally performed on formalin-fixed,
increased dramatically in veterinary medicine over the past paraffin-embedded tissue specimens. Specimens that have been
10 years. Because of the exquisite sensitivity of some assays, espe- archived for several years may still be adequate, even for detec-
cially those that involve DNA amplification, positive results may tion of RNA, which is more labile than DNA.6,7 The optimum
reflect contamination that occurs in the laboratory. Contamina- specimen for fluorescence in situ hybridization (see later for dis-
tion problems have decreased with increased automation and cussion) is tissue that has been snap frozen in liquid nitrogen
the use of real-time PCR assays (see later). In human medicine, after being embedded in optimal cutting temperature (OCT)
proficiency testing programs have helped to overcome quality medium.5
assurance problems in laboratories that perform molecular diag-
nostic assays.2 Not all PCR assays for a specific pathogen are equal, Polymerase Chain Reaction
because the nucleic acid primers and probes and the equipment When collecting specimens for PCR, the timing of collection rela-
used to run each assay may be different. So a PCR assay for Lepto- tive to the course of disease and the best specimen type must be
spira performed in one laboratory may perform differently than an considered. For acute diseases, PCR is often most useful early in
assay used in another laboratory. the course of disease. For chronic diseases, timing is less critical.

37
38 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

A B
FIGURE 5-1  Fluorescence in situ hybridization (FISH). A, A nucleic acid probe that is complementary to a segment of leptospiral DNA is tagged with a fluorescent label and incubated
with a tissue section on a slide. B, Unbound probe is washed away, and the section is examined using fluorescence microscopy. (Leptospira FISH image courtesy Dr. Richard Goldstein.)

BOX 5-1
Applications of Nucleic Acid Based Assays

Application Examples
Detection of microbes that are not easily detected using Bartonella spp., viral pathogens
light microscopic examination of tissues or smears
Detection of microbes that are difficult or impossible to Hemotropic mycoplasmas, Bartonella spp., Mycobacterium
culture spp., Brucella spp.
Rapid detection of microbes to allow early institution of Influenza virus, hypervirulent FCV, rabies virus, Mycobacte-
infection control measures rium tuberculosis
Detection of microbes that are hazardous to culture in the H1N1 influenza virus, Francisella tularensis
laboratory
Detection of microbes in the face of inactivating substances Retrospective identification of an infecting microorganism in
such as antimicrobials or formalin formalin-fixed, paraffin-embedded tissues after histopathology
Detection of antimicrobial resistance genes Possession of mecA, the gene for methicillin resistance, by
staphylococci
Detection of novel pathogens using broad-spectrum PCR Bartonella henselae,* hemotropic mycoplasmas†
and sequencing
Monitoring organism load as a measure of disease progres- FIV viral load
sion or response to treatment
Identification of a cultured organism to the species level Nocardia and Actinomyces spp., many molds

FCV, Feline calicivirus.


*Anderson B, Sims K, Regnery R, et al. Detection of Rochalimaea henselae DNA in specimens from cat scratch disease patients by PCR. J Clin
Microbiol 1994;32:942-948.
†Willi B, Boretti FS, Cattori V, et al. Identification, molecular characterization, and experimental transmission of a new hemoplasma isolate from a

cat with hemolytic anemia in Switzerland. J Clin Microbiol 2005;43:2581-2585.

Knowledge of organism shedding patterns can help to determine should be submitted. In general, DNA is stable in tissue for up
the most appropriate timing of specimen collection. For example, to 24 hours at 2°C to 8°C, at least 2 weeks at −20°C, and at
Leptospira organisms are primarily present in blood in the first least 2 years at −20°C or below −70°C.5 Specimens held at room
week of acute illness, after which they may be shed in the urine.8 temperature should be submitted immediately and reach the
Because contamination can occur outside the PCR labora- laboratory within 24 hours. Specimens can also be refrigerated
tory as well as within the laboratory, collection of specimens and submitted on wet ice within 72 hours. Provided the target
for nucleic acid testing should be performed aseptically. Gloves of the assay is not an erythrocyte pathogen, erythrocytes should
should be worn, and disposable instruments should be used be removed before storage if possible. Specimens should not be
(e.g., disposable blades, punch biopsy instruments). stored in frost-free freezers, as these undergo repeated freeze-
In general, for detection of pathogens that contain DNA (i.e., thaw cycling, which can be associated with DNA degradation.9
all bacteria, fungi, some viruses), either fresh or frozen specimens Box 5-2 outlines recommendations based on specimen type.
CHAPTER 5  Nucleic Acid Detection Assays 39

BOX 5-2
Guidelines for Collection and Transport of Specimens for Polymerase Chain Reaction Assays

Specimen Type Collection and Transport Recommendations


Whole blood and Use only EDTA or acid citrate dextrose (ACD) anticoagulant. Heparin inhibits PCR.* The use of hemolyzed
bone marrow specimens and frozen blood should be avoided if possible.
DNA: Submit specimens held at room temperature within 24 hours of collection. Specimens can be stored at
2°C to 8°C for 72 hours, or at −20°C or ≤−70°C for prolonged storage (removal of erythrocytes recommended).
RNA: Use stabilization solution or send immediately to the laboratory on wet ice. For storage, freeze
serum, plasma, or buffy coat specimens at or below −70°C.
Tissue specimens Optimal specimen is 1 to 2 g of fresh tissue. Rinse blood from tissue using sterile saline. Wrap in saline-
moistened gauze or paper. Avoid excessive dilution. Formalin-fixed, paraffin-embedded tissues can be used
when no other specimen is available but are not optimal.
DNA: Transport fresh tissue immediately to the laboratory on wet ice. Any storage should be at or below
−70°C, preferably after snap freezing in liquid nitrogen.
RNA: Use stabilizing solution, process within 1 hour of collection, or snap freeze in liquid nitrogen and
store at or below −70°C
Cerebrospinal DNA: Transport at 2°C to 8°C or freeze at −20°C or at or below −70°C
fluid RNA: Chill immediately on wet ice and deliver to the laboratory within 4 hours. Otherwise freeze ­immediately
(removal of contaminating erythrocytes recommended) and ship on dry ice.
Bronchoalveolar Transport at room temperature within 24 hours of collection. If a delay is anticipated, store for up to
lavage specimens 72 hours at 2°C to 8°C or freeze at or below −70°C.
Urine Minimize storage at room temperature. Storage and transport should be according to the specific assay performed.
Feces Use preserved specimen transport container, or transport under refrigeration at 2°C to 8°C.

Adapted from the CLSI guidelines MM13-A.


*Beutler E, Gelbart T, Kuhl W. Interference of heparin with the polymerase chain reaction. Biotechniques 1990;9:166.

RNA is highly susceptible to degradation during storage and Some of these can also be used to detect the same pathogens
transport. For detection of RNA viruses, some laboratories pro- in specimens from dogs or cats. Probes are available for use in
vide an RNA stabilizing solution into which specimens are col- the clinical microbiology laboratory to identify organisms that
lected to prevent RNA degradation. If stabilizing solution is not have grown in culture (e.g., Gen-Probe Inc., San Diego), and
readily available at the time of specimen collection, swabs and these probes are widely used in human clinical microbiology
body fluids should be immediately sent to the laboratory on wet laboratories.
ice. Tissue should ideally be snap frozen at −70°C within half an Probe hybridization can be performed on a nitrocellulose
hour of collection and shipped on dry ice without being thawed in membrane (solid-phase hybridization); on formalin-fixed,
the interim. RNA is stable for at least 2 years at or below −70°C. paraffin-embedded sections mounted on a microscope slide
Ribonucleases can continue to degrade RNA at −20°C. (in situ hybridization); or in solution (liquid-phase hybridiza-
For tissue specimens, the optimal amount is 1 to 2 g, although tion). For solid-phase hybridization, the probe is reacted with
more or less may be required depending on the cellularity of the microorganism DNA that has been immobilized on the mem-
specimen.5 Tissues stored in formalin, especially for prolonged brane. Unbound probe is washed away, and the bound probe
periods, are suboptimal for PCR because the formalin cross- is detected using fluorescence, chemiluminescence, radioactivity,
links the DNA in the specimen. Tissues fixed briefly in formalin or color development (in the same way that bound antibody or
and then paraffin-embedded are preferable to tissues stored in antigen is detected in an ELISA or immunofluorescent antibody
formalin, because the formalin is removed during the embed- assay). For in situ hybridization, formalin-fixed, paraffin-embed-
ding process. Paraffin-embedded specimens should only be used ded specimens are sectioned and mounted on a special slide. The
when no other specimens are available.5 sections are deparaffinized, dried, and incubated with a solution
Although PCR can be performed on feces, its sensitivity may that contains the probe, so both the presence and the location
be lower than with other specimens because inhibitors of PCR of the target pathogen within tissues can be identified. In situ
are particularly abundant in fecal material.10 The use of appro- hybridization assays that include a fluorescent-labeled probe are
priate amplification controls by the laboratory facilitates detec- referred to as fluorescence in situ hybridization (FISH) assays
tion of false negatives due to the presence of inhibitors. (see Figure 5-1).
Because liquid-phase hybridization occurs in solution,
Diagnostic Methods unbound probe cannot be washed away. To overcome this
problem, a chemiluminescent acridinium ester label is attached
Nucleic Acid Probes to the probe. A subsequent chemical hydrolysis step selectively
A variety of nucleic acid probes are available commercially degrades only unbound probe. On addition of peroxides, the
as kits for detection of microorganisms that infect humans. intact (hybridized) probe then emits light.11
40 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

Although not yet widely used for veterinary applications, roughly 20 nucleotides long, is then used to bracket a desired
peptide nucleic acid (PNA) probes are now increasingly avail- DNA sequence, which is subsequently copied using a DNA
able to detect target DNA. PNA probes are uncharged peptides polymerase enzyme (Figure 5-2).
that mimic DNA and bind to complementary DNA sequences PCR assays are broadly divided into conventional PCR
just as a nucleic acid probe would.12,13 PNA probes lack the assays and real-time PCR assays. Conventional PCR assay is
net negative charge of nucleic acid probes; therefore, the elec- the simplest PCR method and although still widely for research
trostatic repulsion that normally occurs when two negatively purposes, its use for diagnostic purposes is decreasing. In con-
charged DNA strands hybridize does not occur. The result is a ventional PCR, the PCR product is detected as a band on an
more stable and specific binding of the probe to its target, which agarose gel after agarose gel electrophoresis. In real-time PCR,
in turn can be associated with increased assay sensitivity and a fluorescent signal is generated as PCR products accumulate,
specificity. allowing them to be detected in “real time.” A major advantage
Branched DNA assays and hybrid capture assays are highly of real-time PCR methods is that the tube in which the PCR
sensitive hybridization methods that include steps to intensify takes place does not need to be opened for detection of the PCR
the signal generated from probe hybridization.3,4 They are not product. This greatly reduces the likelihood of contamination
yet widely used in veterinary medicine. and false positive test results. Real-time PCR assays are ame-
nable to automation, and reactions are generally loaded onto
The Polymerase Chain Reaction covered 96- or 384-well plates. This can be done robotically,
PCR allows the specific amplification of DNA sequences from further reducing the possibility of contamination and manual
just one copy to millions of copies, which can be more readily loading errors.
detected. The DNA from a clinical specimen is extracted using a Regardless of the type of PCR assay, extracted DNA is added
commercially available DNA extraction kit. A pair of primers, to aliquots of a PCR mastermix. The mastermix is a solution

A C
B
D

FIGURE 5-2  Basic PCR assay. Nucleic acid is extracted from a clinical specimen through removal of contaminating proteins such as hemoglobin. Two primers are added that hybridize
at each end of a specific segment of a pathogen’s DNA. Also present in the reaction mixture are the enzyme Taq polymerase (dots) and free nucleotides (dNTPs) for new DNA. A thermocy-
cler is used to rapidly and repeatedly heat and cool the tubes through denaturation, annealing, and extension steps. Denaturation separates double-stranded DNA. Annealing occurs at a
specific temperature that causes the primers to bind only to their target sequences. When the reaction mixture is then heated to 72°C, the Taq enzyme uses the primers as initiation points
for DNA extension, and the target sequence is copied. The process is repeated 30 to 50 times, with logarithmic accumulation of the PCR product, so there are millions of copies of the target
pathogen’s DNA. These can then be detected using agarose gel electrophoresis (see Figure 5-3).
CHAPTER 5  Nucleic Acid Detection Assays 41

that contains the two primers, dNTPs (G, T, A, and C) for mak- Two major types of real-time PCR assays are in use in veteri-
ing new DNA, and a DNA polymerase. The DNA polymerase nary medicine: fluorescent probe–based assays and SYBR Green
used most commonly (Taq DNA polymerase) is a heat-stable PCR assays.
polymerase derived from Thermophilus aquaticus, a bacterium Fluorescent probe real-time PCR assays incorporate one or
that is adapted to live in hot springs. Typical PCR assay reaction more specific DNA probes into the PCR mastermix, in addition
volumes are 10 to 50 µL. to the two primers used in conventional PCR. The probe(s) are
The next step is performed in a thermocycler, a machine that tagged with a dye that emits fluorescent light. Inclusion of a
is programmed to heat and cool the tubes repeatedly, so that probe adds specificity to the test, because it is less likely that
the DNA can be amplified (see Figure 5-2). Initially the DNA both primers and the probe will bind to DNA other than that of
is heated to 95°C, which causes the paired strands of DNA to the target organism. Three types of probes may be used:
denature. Next, the tubes are cooled to a specific annealing 1. TaqMan (5′ nuclease) probes. Many veterinary laborato-
temperature (usually around 50°C to 65°C), which allows the ries that offer probe-based PCR assays use TaqMan probes.
primers bind to their respective sequences. Finally, the tubes are These are tagged with a fluorescent reporter dye at one end,
heated to 72°C. This is the temperature at which the DNA poly- and a quencher dye at the other end of the probe. The close
merase adds dNTPs in the mastermix to the primers, extending proximity of the quencher dye to the reporter dye inhibits
the primers as it copies the sequence of the template DNA. The the reporter dye from emitting fluorescence. When the Taq
3-step cycle is repeated 30 to 50 times, which results in loga- DNA polymerase copies the section of DNA between the
rithmic accumulation of a PCR product that is an exact copy two primers, it encounters the probe and chops it into pieces,
of the segment of DNA between the two primers. The amplified because it has inherent exonuclease activity. This causes the
segments of DNA are called amplicons. reporter dye to separate from the quencher dye. The reporter
dye then emits fluorescence, which can be measured using a
Detection of the PCR Product: Conventional versus Real-time PCR spectrophotometer (Figure 5-4, A).
In conventional PCR, amplified DNA is loaded into wells in an 2. Molecular beacons. A molecular beacon is a probe with fluo-
agarose gel then passed through the gel using horizontal electro- rescent reporter and quencher dyes at each end, but when
phoresis, stained with a DNA stain, and visualized using ultra- not bound to the target sequence, it forms a hairpin loop
violet transillumination. The longer the PCR product, the more that brings each dye in close proximity and stops the reporter
slowly it moves through the gel during electrophoresis. When dye from emitting fluorescence. When the probe anneals, the
a series of molecular weight markers are run alongside the end dyes separate, and the reporter dye emits fluorescence (see
product of PCR, the size of the PCR product can be estimated Figure 5-4, B).
(Figure 5-3). The true identity of the product can then be con- 3. FRET probes. FRET (fluorescent resonance energy transfer)
firmed using sequencing or by transferring it to a membrane probe assays use two probes that are expected to lie next
and performing DNA hybridization with a specific probe. The to one another on the target sequence. One of the probes is
process of loading the PCR product in agarose wells requires tagged at the 3′ end with a green fluorescent dye, the other
that the PCR tubes be opened, which can lead to aerosoliza- at the 5′ end with a red fluorescent dye. When the two dyes
tion of PCR amplicons. These amplicons can then contaminate come together during annealing, the green fluorescent dye
reagents, equipment, and tubes that are used for running sub- transfers its energy to the red probe, causing it to emit flu-
sequent PCR assays. Unless proper controls are included, false orescence, which can then be detected (see Figure 5-4, C).
positives resulting from such contamination may go unidenti- Currently there are only rare reports of the use of this tech-
fied and lead to erroneous reporting of results. nology for diagnosis in veterinary medicine.

A B
FIGURE 5-3  A, Detection of PCR products by agarose gel electrophoresis. At the end of the PCR process, the reaction mixture that results is loaded into wells at the top of an agarose
gel. An electric current is applied across the gel, and because DNA is negatively charged, any amplified DNA in the reaction mixture migrates through the gel toward the positive terminal.
The longer the DNA sequence that has been amplified, the more slowly it moves through the gel. Simultaneous electrophoresis of a molecular weight standard in an adjacent lane (left
lane) allows estimation of the size of any PCR products amplified. The molecular weight standard contains DNA fragments of known size. After electrophoresis, the gel is stained with a DNA
stain. If the target pathogen’s DNA was successfully amplified, it appears as a band of an expected size (in this case, 485 base pairs [bp] long). Bands that are the wrong size (“aberrant PCR
products”) may represent nonspecific amplification of DNA from a different microorganism or host DNA (middle lane). B, Photograph of PCR products in an agarose gel after agarose gel
electrophoresis and staining. DNA bands amplified from Chlamydia felis, feline calicivirus (FCV), and feline herpesvirus-1 (FHV-1) are shown. The primers were designed to amplify different-
length products from each pathogen so that if multiple pathogens were present in a single specimen, they could be distinguished from one another.
42 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

A B

C
FIGURE 5-4  Probe-based real-time PCR assays. A, TaqMan probe PCR system. In addition to the forward and reverse primers used in conventional PCR, an additional short probe is used
that binds specifically to the DNA of the pathogen of interest (step one). Emission of fluorescence by a reporter dye located at one end of the probe is quenched by another dye at the other
end of the probe. When the Taq enzyme extends the primers, an inherent exonuclease activity of the enzyme results in the digestion of the probe. The quencher and reporter dyes separate,
causing the quencher dye to emit fluorescence. Other probe-based real-time PCR assays include molecular beacon assays (B) and fluorescent resonance energy transfer probe assays (C).

SYBR Green Real-time PCR assays use a double-stranded DNA positive. Said another way, if PCR proceeds to 40 cycles with-
binding dye (SYBR Green) that fluoresces when bound to double-­ out emission of fluorescence (CT = 40), then the target DNA is
stranded DNA as it forms during PCR amplification. Because not detectable in the specimen, and the result is reported as nega-
there is no probe, carefully designed primers are required to tive. The lower the CT value, the more target DNA (= organism
ensure high assay specificity for the organism of interest. load) is present in the sample, because fewer cycles elapse before
The amount of fluorescence emitted during real-time PCR is fluorescence is detected. Because specimen size can vary, the CT
proportional to the number of copies of template DNA in the value may be normalized to the number of host cells present in
specimen, so real-time PCR assays can be quantitative. A plot of the sample. Knowledge of organism load may have prognostic
PCR cycle number against fluorescence is generated by an attached value for some infections, can allow infection to be monitored
computer and interpreted by the laboratory technician (Figure over time with treatment, and may be useful to differentiate low-
5-5). The number of cycles that elapse before fluorescence is level colonization from active infection causing clinical signs (see
detectable above a certain threshold value is the CT (cycle thresh- Interpretation of Results, later). At the time of writing, organism
old) value. Most laboratories run approximately 40 cycles, and loads are variably reported by veterinary diagnostic laboratories
so specimens that generate CT values less than 40 are considered performing PCR assays.
CHAPTER 5  Nucleic Acid Detection Assays 43

FIGURE 5-5  Plot that shows the emission of fluorescence during PCR product formation over time in real-time PCR as the number of cycles of denaturation, annealing, and extension
increases from 0 to 40 (x-axis). The cycle threshold (CT) value is the number of PCR cycles required for the fluorescent signal (y-axis) to rise above a threshold value for fluorescence (threshold
line). The more copies of target pathogen DNA that are present in a clinical specimen (which correlates with the number of organisms present), the earlier fluorescence is emitted and the
lower the CT value. Thus, for example, if only one organism is present in a submitted specimen, the CT value for this particular PCR assay is 32, but if 10,000 organisms are present, the CT
value is around 19.

FIGURE 5-6  Nested PCR. After the first round of PCR amplification, a second set of internal primers (green) is used in yet another round of PCR amplification, which generates a second,
shorter PCR product.

Rapid thermocyclers are available in several different for- complementary DNA primer that binds to the RNA, or alterna-
mats. Special carousel thermocyclers have been developed that tively, random hexamers, which are a mixture of six-nucleotide
rapidly heat and cool the reactions, which are placed in specially primers that represent all possible combinations of dNTPs. The
designed glass capillaries. As a result, PCR assays can be com- reverse transcriptase then uses the primers to make a DNA copy
pleted in 45 minutes, instead of the usual 4 to 5 hours.14 of the RNA (known as cDNA), which is subsequently subjected
to PCR amplification.
Polymerase Chain Reaction Variations For nested PCR, a small amount of amplified PCR product is
Variations on the basic PCR assay include reverse-transcriptase subjected to a second round of conventional PCR using another
PCR, nested PCR, and multiplex PCR. set of internal primers (Figure 5-6). Having an additional set
Reverse-transcriptase PCR is used for RNA detection. After of primers increases the specificity of the PCR assay and also
extraction of RNA from the clinical specimen using a com- greatly increases its sensitivity.15 Unfortunately, the increased
mercial kit, the enzyme reverse transcriptase is added to the sensitivity means that false positives due to contamination can
RNA, together with dNTPs that will be used to make DNA. be extremely problematic with this method, and it has lost pop-
The starting point for reverse transcription can be a single ularity for diagnostic purposes.
44 SECTION 1  Laboratory Diagnosis of Canine and Feline Infectious Diseases

Multiplex PCR assays use multiple sets of primers in the same


reaction tube to simultaneously detect multiple organisms (or mul- BOX 5-3
tiple genes from the same organism). Duplex PCR assays can detect
two organisms, and triplex PCR assays can detect three organisms.
Methods That May Be Used to Minimize Contamination
The potential downside of multiplex reactions is that if both organ- in Diagnostic Laboratories That Perform Nucleic Acid
isms are present, and one organism is present in relative abun- Amplification Assay
dance, amplification of this organism can consume reagents, which
reduces the sensitivity of the assay for the other target organism. Separation of rooms for DNA extraction, PCR, and PCR
product detection
Verification of Polymerase Chain Reaction Assay Performance Robotic DNA extraction
Before being used for diagnostic purposes, PCR assays must be Automated detection of PCR products (real-time PCR)
shown to be sensitive, specific, reproducible, and efficient. Opening one specimen tube at a time
For PCR, the limit of detection is the lowest number of Use of screw-top caps rather than snap-top tubes to pre-
copies of the target DNA or RNA sequence that the assay can vent splashing
detect. Serial dilutions of known amounts of the template DNA Use of filter-plugged pipette tips
are tested using the PCR assay of interest, until template can no Use of negative extraction controls and negative template
longer be detected. controls in every run
From a diagnostic standpoint, a PCR assay may lack sensi- Routine monitoring for contamination using wipes
tivity (many false negatives) if the assay fails to detect all pos- Standard cleanup protocol that inactivates nucleic acids
sible strains of a target organism. This can occur if the probe or Use of positive control samples of low concentration
primers are strain, rather than species-specific, and many strains Use of dUTP and uracil-N-glycosylase in the PCR
of the target organism exist with variable sequences (as is the mastermix
case with RNA pathogens such as feline calicivirus). As part of
the verification process, the assay should be shown to detect as
many strains as possible. process to ensure that contamination does not occur during
A PCR assay has poor analytical specificity if false positives extraction (negative extraction control). When 96- or 384-well
occur as a result of contamination or because of manual loading plate formats are used, negative control wells are interspersed
error. Specificity may also be poor if the primers bind to and ini- among the test wells to monitor for plate contamination.
tiate amplification of DNA from unrelated organisms or the host In order to prevent contamination, some laboratories use
animal (i.e., cat, dog, human DNA). Specificity is tested during deoxyuracil triphosphate (dUTP) instead of deoxythymidine
the verification process by showing that the PCR assay fails to triphosphate (dTTP) in the PCR mastermix. The PCR products
amplify DNA from specimens that contain organisms that are therefore contain uracil (U) in place of thymidine (T). The enzyme
genetically related to the target organism, as well as organisms uracil-N-glycosylase (UNG) is also added to the mix. This enzyme
that may be expected to be contaminating normal flora. destroys DNA amplicons that contain deoxyuracil residues, and
The results of PCR assays must also be repeatable. Repeatabil- eliminates any contaminating DNA amplicons from previous
ity is determined for real-time PCR assays by determining whether reactions before PCR amplification begins. When the specimen
the CT value varies significantly for the same specimen when is heated in the initial denaturation step, the UNG is destroyed.
tested multiple times. Usually intraassay (within the same run) The presence of substances that inhibit the PCR assay can
and interassay (within different runs) variations are measured. be detected using a housekeeping gene control PCR assay. This
A real-time PCR assay that has an efficiency of 100% doubles is performed on a separate aliquot of the specimen at the same
the number of copies of template in every PCR cycle. Efficiencies time as the assay for the target pathogen. Housekeeping genes
below 90% are unacceptable for quantitative assays, and ideally are host (dog or cat) genes that are expected to be present in the
efficiency should be greater than 95%. The efficiency is deter- specimen. Examples of commonly used housekeeping gene PCR
mined by testing serial dilutions of a target DNA standard and assays are those for canine or feline glyceraldehyde-3-phosphate
by creating a chart of copy number versus CT value. Ideally, this dehydrogenase (GAPDH), histone, or 18S DNA. A negative
is compared with a similar chart using dilutions of the standard housekeeping gene PCR assay result (or a high CT value for the
DNA spiked into a clinical specimen, to show that inhibitors assay, which is equivalent to a weak positive result) suggests
within the specimen will not affect efficiency. that inhibitors may be present in the specimen, or that the DNA
in the submitted specimen may have degraded during storage or
Prevention of Contamination and Quality Control transport. Some PCR assays use an internal housekeeping gene
Prevention of PCR contamination relies on good laboratory control, in which a housekeeping gene PCR is multiplexed with
practices, some of which are outlined in Box 5-3. Contamination the PCR assay for the target microorganism.
can be difficult to eradicate once it occurs, so prevention of con-
tamination is critical. Laboratories should include negative con- Interpretation of Results
trols with every run, and a positive control should be included
to ensure that the assay is working, preferably one that is put Because some PCR assays can detect as few as one or two organ-
through the nucleic acid extraction process. The positive con- isms, and both viable and nonviable organisms are detected, it
trol should contain a low concentration of DNA, as abundant may be difficult to determine the significance of a positive PCR
DNA in a positive control sample may be a source of contamina- test result in light of an animal’s disease. An understanding of
tion. Many laboratories run two separate negative controls. One the pathogenesis of infectious diseases and organism shedding
of the negative controls contains mastermix and nuclease-free patterns is crucial in interpreting positive PCR results. Correct
water in place of specimen DNA. The other consists of water or interpretation of the results relies on knowledge of the methods
a known negative specimen that is placed through the extraction used by a laboratory and their advantages and disadvantages.
CHAPTER 5  Nucleic Acid Detection Assays 45

A positive result can occur if an attenuated live vaccine for


BOX 5-4 the target organism was recently administered. Because PCR
detects both viable and nonviable organisms, PCR can be posi-
Factors That Should Be Considered When Interpreting tive for days after antimicrobials have been administered, when
Nucleic Acid Amplification Assay Results culture is negative.17-19 The duration of PCR positivity after
treatment depends on the specimen type tested and the target
Negative Test Result pathogen.
Organism not present
Too few organisms present REFERENCES
Insufficient specimen size 1. Komminoth P, Werner M. Target and signal amplification:
Wrong specimen type approaches to increase the sensitivity of in situ hybridization. His-
Improper timing of specimen collection tochem Cell Biol. 1997;108:325-333.
Patient receiving antimicrobial therapy 2. Quality Control for Molecular Diagnostics. 2012. http://www.qcmd.
Specimen degradation during storage or transport org/. Last accessed May 11, 2012.
Assay reagent failure 3. Andras SC, Power JB, Cocking EC, et al. Strategies for signal ampli-
Assay inhibitors present or extraction failure fication in nucleic acid detection. Mol Biotechnol. 2001;19:29-44.
Reagents consumed in a multiplex assay 4. Nolte FS, Caliendo AM. Molecular microbiology. In: Versalovic J,
Carroll KC, Funke G, et al. eds. Manual of Clinical Microbiology.
Assay does not detect all strains
10th ed. Washington, DC: ASM Press; 2011:27-59.
5. Clinical and Laboratory Standards Institute. Collection, transport,
Positive Test Result preparation, and storage of specimens for molecular methods;
Viable or nonviable organisms present and causing disease approved guideline. MM13-A. 2005;25:7-18.
Viable or nonviable organisms present but not causing 6. Illig R, Fritsch H, Schwarzer C. Breaking the seals: efficient mRNA
disease detection from human archival paraffin-embedded tissue. RNA.
Vaccine organisms present 2009;15:1588-1596.
Contamination 7. Tan LH, Do E, Chong SM, et al. Detection of ALK gene rearrange-
Loading errors ments in formalin-fixed, paraffin-embedded tissue using a fluorescence
Assay cross-reaction with other pathogens or host DNA in in situ hybridization (FISH) probe: a search for optimum conditions of
tissue archiving and preparation for FISH. Mol Diagn. 2003;7:27-33.
the sample
8. Greenlee JJ, Bolin CA, Alt DP, et  al. Clinical and pathologic
comparison of acute leptospirosis in dogs caused by two strains
of Leptospira kirschneri serovar grippotyphosa. Am J Vet Res.
2004;65:1100-1107.
Specimens That Test Negative 9. Lahiri DK, Schnabel B. DNA isolation by a rapid method from
Factors that lead to negative test results are shown in Box 5-4. human blood samples: effects of MgCl2, EDTA, storage time, and
An example of the use of the wrong specimen type would be sub- temperature on DNA yield and quality. Biochem Genet. 1993;31:
mission of blood for a Borrelia burgdorferi PCR test, when B. 321-328.
burgdorferi is found predominantly in connective tissue.16 Assay 10. Oikarinen S, Tauriainen S, Viskari H, et al. PCR inhibition in stool
samples in relation to age of infants. J Clin Virol. 2009;44:211-214.
inhibition and specimen degradation can be identified by interpre-
11. Arnold Jr LJ, Hammond PW, Wiese WA, et  al. Assay for-
tation of housekeeping gene control assay results, which should
mats involving acridinium-ester-labeled DNA probes. Clin Chem.
be performed by the laboratory. Assay reagent failure is identified 1989;35:1588-1594.
by the laboratory when the positive control tests negative. 12. Padilla E, Manterola JM, Rasmussen OF, et  al. Evaluation of a
fluorescence hybridisation assay using peptide nucleic acid probes
Specimens That Test Positive for identification and differentiation of tuberculous and non-­
Factors that must be considered when interpreting positive test tuberculous mycobacteria in liquid cultures. Eur J Clin Microbiol
results are shown in Box 5-4. The most common reason for false- Infect Dis. 2000;19:140-145.
positive test results for assays that involve DNA amplification is 13. Zhang N, Appella DH. Advantages of peptide nucleic acids as diag-
contamination. Loading errors, in which specimens are inadver- nostic platforms for detection of nucleic acids in resource-limited
settings. J Infect Dis. 2010;201(suppl 1):S42-S45.
tently loaded into the wrong wells on a microtiter plate, may also
14. Cockerill 3rd FR. Application of rapid-cycle real-time polymerase
lead to false-positive results and are reduced with the use of auto-
chain reaction for diagnostic testing in the clinical microbiology
mated methods. Because of the exquisite sensitivity of PCR, it may laboratory. Arch Pathol Lab Med. 2003;127:1112-1120.
be difficult to determine the significance of a true positive result. 15. Lu JJ, Chen CH, Bartlett MS, et  al. Comparison of six different
The organism may be part of the normal flora, carried subclinically PCR methods for detection of Pneumocystis carinii. J Clin Microbiol.
(e.g., latent feline herpesvirus-1 in a nasal biopsy), or simply colo- 1995;33:2785-2788.
nizing the anatomic site sampled. Several U.S. veterinary laborato- 16. Aguero-Rosenfeld ME, Wang G, Schwartz I, et  al. Diagnosis of
ries offer PCR “panels” for diagnostic purposes. Individual PCR Lyme borreliosis. Clin Microbiol Rev. 2005;18:484-509.
assays for several different pathogens are performed on nucleic 17. Bockenstedt LK, Mao J, Hodzic E, et al. Detection of attenuated,
acid extracted from a single specimen. This high-throughput panel noninfectious spirochetes in Borrelia burgdorferi-infected mice
after antibiotic treatment. J Infect Dis. 2002;186:1430-1437.
format allows assays to be offered at a substantial discount over
18. Braddock JA, Tasker S, Malik R. The use of real-time PCR in the
the cost of multiple individual assays. This type of testing can result
diagnosis and monitoring of Mycoplasma haemofelis copy number in a
in unexpected positive test results, such as a positive result for the naturally infected cat. J Feline Med Surg. 2004;6:161-165.
Clostridium perfringens toxin gene in a cat with diarrhea that is 19. Sykes JE, Studdert VP, Browning GF. Comparison of the poly-
suspected to have trichomoniasis. As with any laboratory test, merase chain reaction and culture for the detection of feline Chla-
positive results need to be interpreted in light of the clinical history, mydia psittaci in untreated and doxycycline-treated experimentally
clinical signs, and the pathogenesis of the disease in question. infected cats. J Vet Intern Med. 1999;13:146-152.
SECTION 2
Antiinfective Therapy
Jane E. Sykes and Mark G. Papich

CHAPTER 6

Principles of Antiinfective Therapy


Jane E. Sykes and Mark G. Papich

w KEY POINTS
• T he use of antimicrobial drugs (AMDs) can lead to selection of organism, (3) selection of agents with activity specific to the patho-
resistant bacterial populations, within the targeted bacterial gen present, (4) administration of an adequate dose of AMD, and
population and also other commensal bacteria. (5) administration of AMD for the proper duration of time.
• Bacterial mechanisms of drug resistance include production of • Selection of an appropriate AMD should be based on the activ-
enzymes that inactivate AMDs, drug exclusion through mem- ity of the agent against the suspected pathogen; knowledge
brane alterations and efflux pumps, and modification of the site of drug pharmacokinetics, which includes bioavailability and
of action of the AMD. tissue penetration; consideration of host factors such as con-
• Strategies that minimize selection for drug resistance include current illness, medications, or immunosuppression; and knowl-
(1) confirmation of the presence of infection before AMD treat- edge of drug pharmacodynamics, that is, whether the drug
ment is commenced, (2) proper identification of the infecting exhibits concentration- or time-dependent antibacterial effects.

INTRODUCTION encouraged to develop antimicrobial guidelines based on evalua-


tion of their local prevalence of AMD resistance. Strategies that mini-
Antibiotics have been considered one of the greatest inventions mize selection for antimicrobial resistance include documentation
of the 20th century. The modern era of antimicrobial drug (AMD) of the presence of infection; proper identification of the infecting
treatment began with the discovery of sulfonamides in 1935.1 In organism; and the use of agents that are as specific for the pathogen
the 1940s, the therapeutic value of penicillin and streptomycin as possible, at the proper dose and for the proper duration of time.
was discovered, and by 1950, the “golden age” of AMD therapy was These strategies are described in more detail in this chapter.
underway. With the increasing use of AMDs, there has been a transi-
tion to an era of widespread antimicrobial resistance among impor- Identification of the Infecting Organism
tant veterinary and human pathogens. This has been compounded
by a declining rate of development of new classes of AMDs. The detection of fever or leukocytosis does not imply bacte-
Antimicrobial resistance genes have existed in microbes long rial infection and the need for antibiotic treatment, especially
before antimicrobial agents were used in therapy. Microbe popula- given the relatively high prevalence of sterile inflammatory and
tions generally consist of a mixture of genetically susceptible and immune-mediated diseases in dogs and viral infections in cats.
resistant organisms. The use of AMDs exerts pressure that favors Selection of the most appropriate AMD relies on knowledge of
selection of the resistant microbes (“survival of the fittest”), within (1) the presence of an infectious agent, (2) the type of infectious
both the targeted bacterial population and other commensal bac- agent present, and (3) the susceptibility of the agent to different
teria (which have the potential to become pathogenic when host AMDs. In some cases, treatment with antibiotics may not be
defenses are impaired). The chance that resistant bacterial popula- necessary even though the cause of disease is bacterial. Some
tions will emerge depends on the ability of the population to rapidly infections undergo spontaneous cures without the aid of an
acquire mutations, the ability of host defenses to eliminate resistant antibiotic. For example, a cat bite abscess can often be drained
bacteria, and the AMD concentrations at the site of infection. and resolved without the need to administer an antibiotic (see
Guidelines for prudent antimicrobial use have been published Chapter 57).
by several professional veterinary organizations in order to reduce In some situations, collection of a specimen for cytology or
selection for resistant bacterial pathogens.2-6 Individual hospitals are histopathology permits a diagnosis of infection to be established.

46
CHAPTER 6  Principles of Antiinfective Therapy 47

The identity of some infectious agents, such as rickettsial and decline in bacterial numbers that occurs when the drug con-
fungal pathogens, may be presumptively identified to the genus centration drops below the minimum inhibitory concentration
or species level based on their morphology. If skin cytology from (MIC) reflects clearance by the host immune response. How-
a dog reveals cocci (a Gram stain is not necessary to confirm the ever, the line that separates bactericidal and bacteriostatic is
presence of gram-positive cocci), then the presence of Staphy- not as precise as previously thought. Some bacteriostatic drugs
lococcus pseudintermedius is highly probable. If, on the other become bactericidal when present in high concentrations at
hand, cytology from an ear swab reveals rods, the possibility of the site of infection, or if concentrations are maintained above
Pseudomonas aeruginosa must be considered. In either case, this the MIC for the entire dosing interval. Even macrolides and
knowledge is valuable for selecting initial antibacterial therapy. chloramphenicol, which are traditionally considered as bac-
The use of molecular diagnostic techniques, such as PCR assays, teriostatic drugs, can be bactericidal against some organisms.
can also allow rapid identification of an infecting organism, A drug may be bactericidal against gram-positive cocci, but
which can help to guide antimicrobial drug selection. For exam- bacteriostatic against gram-negative bacilli. Therefore, from a
ple, identification of a vector-borne pathogen (e.g., Rickettsia clinical perspective, it is more useful to describe drugs as time
spp., Ehrlichia canis) is helpful because doxycycline is usually the versus concentration-dependent, rather than as bactericidal or
first drug of choice for the majority of these pathogens. Unfortu- bacteriostatic. This allows the clinician to consider the dosage
nately, unless susceptibility to a particular AMD is predictable, regimen and dosing interval to optimize therapy. There are rare
these techniques do not typically provide information about instances where a bactericidal agent is preferred over a bacte-
AMD susceptibility. Given the emergence of widespread AMD riostatic one. It may be preferable to prescribe a bactericidal
resistance, especially among bacteria and some fungi, collection drug to treat life-threatening infections (such as vegetations in
of specimens for culture and susceptibility before treatment with endocarditis or in a systemically immunocompromised host).7
AMDs is commenced has become more and more important. Antibacterial drugs have also been classified for veterinary
Although there is an initial cost to the client for culture and sus- use as first-line, second-line, or third-line drugs.8 First-line
ceptibility testing, long-term costs to the client may be reduced drugs are those that could be used for empirical selection in
considerably because of decreased prescription of unnecessary or the absence of or pending the results of culture and suscepti-
inappropriate AMDs, improved likelihood of rapid resolution bility testing, and include amoxicillin, cephalexin, doxycycline,
of the underlying disorder with early diagnosis and appropri- and trimethoprim-sulfonamides. Second-line drugs are those to
ate treatment, and reduced need for more expensive, second- or be used on the basis of culture and susceptibility testing and
third-line AMDs. Administration of AMDs without identifica- because of the lack of any appropriate first-line options. These
tion of the infectious agent can also decrease subsequent chances include ticarcillin, piperacillin, amikacin, and third-generation
of successful culture and proper diagnosis if the disorder does cephalosporins. Fluoroquinolones were also included in this
not resolve. group because in human medicine, excessive fluoroquinolone
In animals with life-threatening infections, institution of use has been associated with emergence of antimicrobial resis-
AMD treatment is necessary before the results of culture and sus- tance and treatment failures.9 Fluoroquinolones could be con-
ceptibility become available. The choice of treatment at this stage sidered as first-line drugs for dogs and cats suspected to have
can be based on cytology and knowledge of the type of infections serious gram-negative bacterial infections that require treatment
that most commonly occur at the anatomic site involved (also pending the results of culture and susceptibility testing. The use
known as bacteriologic statistics). Because of the life-threatening of third-line drugs, including vancomycin, linezolid, and car-
nature of these infections, the practice of “de-escalation” (as bapenems such as imipenem and meropenem, is usually reserved
opposed to “escalation”) is employed. This is the practice of ini- for situations when certain criteria are met8:
tially selecting a highly active agent that has a high probability of 1. Infection must be documented based on clinical abnormali-
successful elimination of the pathogen; then once the infectious ties and culture.
agent has been identified, the drug spectrum should be changed 2. The infection is serious and has the potential to be life-
or narrowed according to culture and susceptibility results. If threatening if left untreated.
the infection is life threatening, highly active agents such as an 3. Resistance is documented to all other reasonable first- and
aminoglycoside or fluoroquinolone with or without a β-lactam second-line options.
can be selected initially. Then, if culture and susceptibility show 4. The infection is potentially treatable.
susceptibility to β-lactams, treatment should be continued with 5. The clinician may seek advice from an infectious disease clini-
these agents alone. cian or a clinical microbiologist to discuss antimicrobial sus-
ceptibility test results, and to discuss the use of these agents if
Classification of Antimicrobial Drugs there is unfamiliarity with their use. In some instances, there
may be other viable options (e.g., topical therapy).
Antimicrobial drugs may be classified based on their struc-
ture (e.g., β-lactams, fluoroquinolones), spectrum of activity Pharmacodynamics of Antimicrobial Drugs
(gram-positive versus gram-negative bacteria), mechanism
of action (Table 6-1, Figure 6-1), whether they are bacterio- Pharmacokinetic and pharmacodynamic parameters (PK-PD
static (inhibit or slow growth) or bactericidal drugs, and their exposure relationships) are important determinants of the effi-
pharmacodynamic properties (see Pharmacodynamics of Anti- cacy of AMDs and serve as the basis for determination of clini-
microbial Drugs, later). Bactericidal drugs interfere with cell cally effective dosage regimens and susceptibility breakpoints
wall or nucleic acid synthesis, whereas bacteriostatic drugs and for the development of guidelines for AMD use for specific
inhibit protein synthesis or cause changes in bacterial physiol- types of infections.10
ogy. When a bacteriostatic drug is removed, in the absence of The killing of microbes by AMDs may be classified as con-
host defense mechanisms, organism growth resumes, and any centration dependent or time dependent. AMDs that exhibit
48 SECTION 2  Antiinfective Therapy

TABLE 6-1
Classification of Antimicrobial Drugs Based on Their Mechanism of Action
Mechanism of Action Antimicrobial Drug Examples
Inhibition of cell wall synthesis β-Lactams, glycopeptides Penicillin, cephalexin, meropenem, vancomycin
Protein synthesis inhibition Tetracyclines, aminoglycosides, chlor- Doxycycline, gentamicin, erythromycin,
amphenicol, lincosamides, macrolides ­clindamycin, azithromycin
Inhibition of DNA replication Fluoroquinolones Enrofloxacin, marbofloxacin
Inhibition of folic acid metabolism Trimethoprim-sulfonamides Trimethoprim-sulfamethoxazole

FIGURE 6-1  Sites of action of antimicrobial drugs within the bacterial cell.

concentration-dependent killing are fluoroquinolones and from irreversible binding of a drug to the target site. The
aminoglycosides. These drugs work optimally when the peak duration of the PAE is influenced by the duration of antibiotic
concentration in the plasma (Cmax), or the area under the exposure, the drug concentration, the bacterial species pres-
curve (AUC) exceeds the MIC by a defined factor (Figure 6-2). ent, and the class of antibiotic. For concentration-dependent
These are typically bactericidal agents and the ability to kill antibiotics, administration of the total daily dose as a single
bacteria increases as the AMD concentrations increase. Target dose every 24 hours is preferred to a smaller divided dose, in
concentrations have been defined for laboratory animals and order to maximize Cmax or AUC. For aminoglycosides, this
humans and have been extrapolated to veterinary patients. also reduces drug toxicity.7
For example, the peak concentration to MIC ratio (Cmax/MIC) β-lactams (penicillins and cephalosporins), macrolides,
should exceed 8 to 10 for optimum dosing of aminoglycosides. and lincosamides (clindamycin) exhibit time-dependent kill-
The AUC for a 24-hour interval to MIC ratio (AUC24/MIC) ing. As concentrations of these drugs increase, bacterial kill-
should exceed 100 for fluoroquinolones. For some infections, ing plateaus, and outcome is correlated with the time that the
a ratio of 125 is better, and in some patients (e.g., immuno- AMD concentration spends above the MIC at the site of infec-
compromised patients with life-threatening infections), a ratio tion. This is usually expressed as the percent of time above the
of 250 may be desirable. Drugs that exhibit concentration- MIC during a 24-hour interval (T > MIC). For β-lactam drugs
dependent killing also have postantibiotic effect (PAE). The apart from carbapenems, PAEs are minimal. As a result, these
PAE is the persistence of antimicrobial effects after drug con- drugs work best when administered multiple times a day or
centrations at the site of infection fall below the MIC. The as continuous-rate infusions, in order to maintain drug con-
exact cause of the PAE varies with the drug, but it may result centrations at the site of infection. For some drugs (e.g., some
CHAPTER 6  Principles of Antiinfective Therapy 49

In animals with renal failure, AMD clearance may be greatly


diminished. In this case, T > MIC and AUC/MIC are achieved
with a reduced dose or less frequent dosing. For β-lactam anti-
biotics, it is logical to increase the dose interval. For fluoroqui-
nolones, it is better to decrease the dose.

Site of Infection
In general, the concentration of an AMD at the site of infec-
tion should at least equal the MIC for the infecting organism,
but the exact concentration and the duration of the concentra-
tion at the site varies with the class of antimicrobial. For some
AMDs, the serum or plasma total (bound and unbound) drug
concentrations may not reflect the drug concentration in tissues.
Examples are the highly lipophilic macrolide antibiotics (e.g.,
azithromycin) for which the tissue drug concentration greatly
overestimates the plasma drug concentration. On the other
hand, tissue concentrations greatly underestimate the plasma or
A serum drug concentration for poorly lipophilic agents such as
aminoglycosides or β-lactams. Antimicrobial selection for dif-
ferent body sites is outlined in Box 6-1.
The concentration of an AMD at the site of infection is
affected by factors such as lipid solubility, protein binding, and
other factors reducing permeability, such as the presence of
large amounts of pus, scar tissue, foreign material, devitalized
tissue, and bone. Lipid-soluble drugs such as chloramphenicol,
rifampin, fluoroquinolones, and trimethoprim are most adept at
crossing membranes, including the blood-brain, alveolar, and
prostatic barriers. In contrast, poorly lipophilic, polar drugs
such as penicillins and aminoglycosides do not cross lipid mem-
branes and will not achieve adequate tissue levels in tissues in
which there is a barrier to penetration (e.g., prostate, brain, eye,
or intracellular sites). In human patients, aminoglycosides and
amphotericin B have been administered intrathecally to treat cen-
tral nervous system infections.11,12 In order to use β-lactam anti-
biotics to treat these infections, high doses are required because
of a poorly penetrated blood-brain barrier. Even in the face of
inflammation, the brain/blood concentration ratio of these drugs
B is low. Water-soluble AMDs that are excreted in active form by
FIGURE 6-2  A, Antimicrobial drug (AMD) pharmacodynamics. Concentration-­ the liver and concentrated in bile, such as doxycycline and ampi-
dependent drugs are most active when the peak concentration (Cmax), or the area-under- cillin, make excellent choices for susceptible hepatobiliary infec-
the-curve (AUC) in the plasma exceeds the MIC by a defined factor. For time-dependent
tions (see Chapter 88). Urinary tract infections are ideally treated
drugs, outcome is correlated with the time that the AMD concentration at the site of
infection spends above the MIC. This is usually expressed as the percent of time above the with drugs such as amoxicillin and trimethoprim-sulfonamides,
MIC during a 24-hour interval (T > MIC). B, As AMD concentrations increase, the ability which are highly concentrated in the urine (see Chapter 89).
of concentration-dependent drugs such as fluoroquinolones (in this case, ciprofloxacin)
to kill bacteria increases. As concentrations of time-dependent drugs such as β-lactam Other Host Factors
drugs (in this case, ticarcillin) increase, bacterial killing plateaus. MIC, minimum inhibitory
concentration. Other host factors that should be considered in AMD selection
and route of administration include the following:
1. History of adverse drug reactions. These are most commonly
cephalosporins), a long T > MIC is achieved by virtue of a long gastrointestinal in dogs and cats, but may include hypersen-
half-life. The required time above the MIC for an AMD varies sitivity reactions.
with the pathogen present, the drug, and the site of infection, 2. Age. With the exception of doxycycline, the use of systemic
but in general it should be 40% to 50% of the dosing inter- tetracyclines is contraindicated in young animals because of
val.7,10 Drug dosages that achieve concentrations in excess of the potential for teeth discoloration (see Figure 8-5). Fluo-
an MIC for these periods can be calculated based on pharma- roquinolones have the potential to cause cartilage and joint
cokinetic data. For time-dependent AMD with long half-lives toxicity in dogs between the ages of 7 and 28 weeks. The
(such as macrolides, clindamycin, and tetracyclines), efficacy is clinical significance of this has been questioned.13,14
determined primarily by the extent to which the area under the 3. Species. Dogs and cats differ in their susceptibility to adverse
curve exceeds the MIC. As the MIC of an organism increases, drug reactions. For example, dogs can develop severe cutane-
it becomes more difficult to achieve these PK-PD targets, and ous reactions to 5-flucytosine. Cats are susceptible to acute
organisms are regarded as resistant. retinal degeneration following treatment with high doses of
50 SECTION 2  Antiinfective Therapy

7. Pregnancy. Penicillins, cephalosporins, aminoglycosides,


BOX 6-1 and macrolides are generally considered safe during preg-
nancy. The volume of distribution in pregnancy is greater,
Examples of Appropriate Initial Antimicrobial Drug Choices and so higher doses may be required to achieve equivalent
for Treatment of Bacterial Infections in Different Anatomic serum concentrations.
Sites* 8. Renal and hepatic function. The dose of AMDs that are
dependent on renal elimination may require reduction in
Skin these animals with impaired renal function in order to mini-
Cephalexin mize toxicity. For example, fluoroquinolones may be more
Amoxicillin-clavulanate likely to cause seizures or retinal toxicity in animals with
Clindamycin renal failure. The use of nephrotoxic drugs such as ampho-
tericin B or aminoglycosides may be relatively contraindi-
Urinary Tract cated in animals with renal dysfunction. Adverse effects may
Amoxicillin be more common when drugs that require hepatic metabo-
Trimethoprim-sulfamethoxazole lism, such as metronidazole or chloramphenicol, are admin-
istered to animals with impaired liver function.
Prostate
Trimethoprim-sulfamethoxazole Route of Administration
Respiratory tract The most common routes of administration for AMDs in dogs and
Doxycycline cats are topical, intravenous, intramuscular, subcutaneous, and
Fluoroquinolones oral. For localized infections of the skin, eye, or ear canal, topical
administration has the advantage of delivering a high concentra-
Intestinal Tract (e.g., for animals with hepatic tion of the AMD to the site, which may overcome bacterial resis-
encephalopathy) tance mechanisms, and avoid any systemic exposure to the drug.
Ampicillin Parenteral administration, especially intravenous administration,
Neomycin provides maximum bioavailability and is recommended for (1)
treatment of life-threatening infections, (2) systemic administration
Biliary Tree of antimicrobials with poor oral bioavailability (such as amino-
Amoxicillin-clavulanate glycosides), and (3) when gastrointestinal signs or malabsorption
Doxycycline preclude effective drug administration via the oral route. Because
Fluoroquinolones parenteral AMDs are generally administered in hospital, compli-
ance with this mode of administration is also likely to be greater.
Brain Absorption may be delayed after subcutaneous or intramuscular
Metronidazole administration when perfusion of these tissues is impaired, and
Fluoroquinolones so these routes are not recommended for use when intravenous
Trimethoprim-sulfonamides administration is possible. Peak plasma drug concentrations are
achieved quickly after intravenous bolus administration. There-
Bone fore, bolus administration is an optimum approach for a concen-
Clindamycin tration-dependent antimicrobial provided rapid injection does not
Cephalosporins produce toxicity (such as may occur with fluoroquinolones). For
Amoxicillin-clavulanate a time-dependent drug such as a β-lactam, a slow infusion (even a
slow constant-rate infusion) optimizes the time-dependent activity.
*The identity of the pathogen present must also be used to guide anti- Provided gastrointestinal function is healthy, administration
microbial drug selection. of many oral AMDs results in acceptable bioavailability, even
though many popular antimicrobials have oral absorption that
is far less than 100%. Drugs with the highest oral bioavailability
enrofloxacin. Cats also are prone to esophageal ulceration in dogs and cats are the fluoroquinolones; β-lactams have low
from oral doxycycline hyclate or clindamycin unless the and variable bioavailability. Nevertheless, once the animal can
medication is administered with a bolus of water or food. tolerate oral medications and its clinical condition becomes sta-
4. Breed. Doberman pinschers may be more susceptible to hyper- ble, a switch can be made from parenteral to oral antimicrobials.
sensitivity reactions following trimethoprim-­sulfonamide Peak plasma concentrations are always lower with oral admin-
administration. istration than those achieved with intravenous administration,
5. Gastric acidity. The absorption of some AMDs, such as but for time-dependent drugs or for drugs that achieve efficacy
ketoconazole and itraconazole, is impaired by medications on the basis of an AUC/MIC exposure relationship, this route is
that suppress gastric acid production. acceptable for a cure. Early switch from intravenous to oral ther-
6. Concurrent medications. Drug interactions, such as concur- apy in human patients has gained increased favor in recent years
rent use of drugs that require metabolism using cytochrome because of reduced lengths of hospitalization, lowered costs,
P450 enzymes, can affect the choice or dose of AMD used. fewer intravascular catheter-related infections, and decreased
For example, chloramphenicol and ketoconazole are well- selective pressure on nosocomial bacterial infections.15,16 The
known P450 enzyme inhibitors, whereas rifampin is an oral bioavailability of some drugs can be maximized if they are
enzyme inducer. administered with food (e.g., clavulanic acid-amoxicillin) or
CHAPTER 6  Principles of Antiinfective Therapy 51

FIGURE 6-3  Genetic mechanisms of resistance in bacteria.

without food (e.g., azithromycin). Some drugs are simply not The single most important mechanism of antibacterial resis-
absorbed orally and must be administered parenterally or by tance is acquisition of foreign DNA by horizontal transfer, espe-
the topical route. These drugs include penicillin G, piperacillin, cially that carried by plasmids and transposons.20 Plasmids are
ticarcillin, aminoglycosides, many cephalosporins (cefazolin, circular, double-stranded DNA molecules that are capable of
cefotaxime, cefoxitin), meropenem, imipenem, and vancomy- independent replication and can be transferred from one bacte-
cin. Some drugs are not absorbed orally unless administered as rial strain or species to another through the process of bacterial
a prodrug. An example is cefpodoxime proxetil. The proxetil conjugation. Transposons are small, mobile segments of DNA
portion of the molecule is cleaved off at the time of intestinal that are flanked by inverted repeats and encode one or more
absorption to allow for adequate systemic concentrations.17 resistance genes (Figure 6-3). They depend on the chromosome
or plasmids for replication. Plasmid-associated genes that con-
Antimicrobial Resistance fer AMD resistance are frequently on transposons and can move
from the plasmid to the bacterial chromosome. A single plasmid
Infectious agents may be intrinsically resistant to an AMD, or may contain resistance genes for more than five different AMDs.
be resistant as a result of genetic variations. Intrinsic or innate
resistance refers to the innate ability of an organism to resist the Mechanisms of Antimicrobial Resistance
effects of an AMD owing to structural or functional characteris- Antimicrobial Inactivation
tics of that organism. For example, enterococci are intrinsically Bacterial enzymes capable of inactivation of AMDs include
resistant to cephalosporins, because they lack the penicillin- β-lactamase enzymes, enzymes that modify aminoglycoside
binding proteins that bind these antibiotics.18 Anaerobic bacte- structure, chloramphenicol acetyltransferase, and erythromycin
ria are intrinsically resistant to aminoglycosides because oxygen esterase. β-Lactamase production is the most important mecha-
is required for entry of the drug to the bacteria. The microbiol- nism of resistance to β-lactam antibiotics among gram-negative
ogy laboratory may “suppress” the results of susceptibility tests bacteria.20 A vast array of β-lactamase enzymes have been dis-
for antimicrobials to which organisms are intrinsically resistant covered, which have been extensively classified into groups.21,22
(i.e., they are not listed in the susceptibility report to the cli- Extended-spectrum β-lactamases (ESBLs) can hydrolyze not
nician). Resistance can also be defined via pharmacokinetic- only penicillins and first-generation cephalosporins, but also
pharmacodynamic targets. For example, an organism may not extended-spectrum cephalosporins such as cefotaxime and cef-
express complete resistance to an antimicrobial, but the organ- tazidime (which contain an oximino group). To date, ESBLs
ism is effectively resistant when standard dosage regimens can- have only been described in gram-negative bacilli and are most
not reach a particular peak concentration or AUC. commonly produced by Escherichia coli and Klebsiella pneu-
Genetic resistance to an AMD may occur as a result of moniae. ESBLs are often inhibited by β-lactamase inhibitors
point mutations, DNA rearrangements, or acquisition of for- such as clavulanic acid. The ESBL with the most importance at
eign DNA. Point mutations can result in alteration of the target the present time for small animals are the CTX-M β-lactamases,
site of an AMD, such as the susceptibility of bacterial DNA which hydrolyze cefotaxime and other third-generation cepha-
gyrase to fluoroquinolones.19 DNA rearrangements may include losporins. Other types of β-lactamase enzymes include AmpC
inversions, deletions, duplications, insertions, or transpositions β-lactamases, which are encoded by chromosomal genes, and
(including chromosome to plasmid) of large segments of DNA. metallo-β-lactamases.20,23 These types of β-lactamase enzymes
52 SECTION 2  Antiinfective Therapy

FIGURE 6-4  Synergistic, antagonistic, and additive effects of antimicrobial drug combinations. Drugs used are referred to using letters of the alphabet. Control = no drug.

are resistant to clavulanic acid. Metallo-β-lactamases can inacti- bacteria, and because of their specificity for certain bacterial spe-
vate carbapenems such as meropenem and imipenem. cies, they can deliver targets to one type of organism without
harming others.27,28 Bacteriophage treatment may also be more
Membrane Alteration successful than AMD treatment in the presence of biofilms, which
Protein channels called porins facilitate the passage of hydro- impair antibiotic penetration. Other approaches that are under
philic AMDs into gram-negative bacteria. Acquisition of porin investigation include use of bacterial cell wall hydrolases, cationic
mutations can result in conformational changes of or reduced antimicrobial peptides, and antisense antibiotics. Cationic anti-
expression of porins. The result is exclusion of AMDs such as microbial peptides are derived from eukaryotic cells, have immu-
β-lactams and fluoroquinolones.24 nomodulatory properties, and rapidly kill a variety of bacteria.29
Although it is difficult for bacteria to develop resistance to these
Efflux Pump Induction peptides, multiple resistance mechanisms have been described.
Expression of efflux pumps by bacteria allows them to exclude Antisense antibiotics are oligonucleotides that bind to the DNA
certain AMDs. This is a major mechanism of resistance to tetra- of pathogenic microorganisms and inhibit gene expression.30
cyclines in gram-negative bacteria. A huge variety of efflux pumps
have been described.25 Efflux pumps may not be specific for one Antimicrobial Drug Combinations
particular class of antimicrobial. Therefore, these have been termed
“multidrug” efflux pumps and mediate multidrug resistance When an infectious agent is suspected but has not yet been identi-
(MDR). The acquisition of MDR mechanisms greatly reduces the fied, the use of AMD combinations is tempting because of the added
effective options for therapy. Multidrug-resistant organisms such broad spectrum of coverage that they provide. However, use of
as Pseudomonas aeruginosa and Acinetobacter baumannii can AMD combinations does not necessarily reduce the chance of selec-
utilize multidrug efflux pumps in concert with a variety of other tion for a resistant organism, adds expense, and may predispose to
resistance mechanisms, including β-lactamase enzyme production, drug toxicity. Theoretically, drug combinations may have additive,
drug target alterations, and AMD modification enzymes. synergistic, or antagonistic effects ­(Figure 6-4). These properties are
not as well established in clinical situations. For example, antago-
Target Modification nism between a bacteriostatic antibiotic (such as tetracycline) and
Methylation of ribosomal targets by bacterial methyltransfer- a bactericidal antibiotic that requires bacterial growth for efficacy
ases can lead to resistance to tetracyclines, lincosamides, ami- (such as a β-lactam) is also theoretically possible, but there is only
noglycosides, and especially macrolide antibiotics. Expression one clinical example of this occurrence, published in the 1950s.31
of altered penicillin binding proteins (PBPs) is an important Such antagonism has not been replicated since that time.
mechanism of resistance to β-lactam antimicrobials. Expres- Several reasons have been suggested for the use of AMD
sion of PBP2a by staphylococci, which is encoded by the mecA combinations:
gene, confers resistance to all β-lactam antimicrobials, including 1. Some drugs must be administered in combination with other
β-lactamase resistant penicillins such as methicillin and oxacillin drugs because of rapid development of resistance to them
(methicillin-resistant staphylococci). Mutations of DNA gyrase when they are used as a sole agent. Examples are 5-flucytosine
can confer resistance to fluoroquinolones.20 Resistance to trim- and rifampin.
ethoprim sulfonamides can result from altered dihydropteroate 2. If a polymicrobial infection is present, one drug may not
synthetase and dihydrofolate reductase enzymes.26 have a sufficient spectrum of activity.
3. If the nature of an infection is not known and infection is
Novel Approaches to Antimicrobial life-threatening, it is reasonable to commence treatment with
Drug Resistance a broad-spectrum combination (such as a fluoroquinolone
With rapid spread of multidrug-resistant bacterial pathogens, and a β-lactam), based on knowledge of the likely pathogen
alternatives to antibiotics have been investigated. One approach present and local prevalence of AMD resistance, pending the
is bacteriophage therapy. Bacteriophages are viruses that infect results of culture and susceptibility testing. Once the results
CHAPTER 6  Principles of Antiinfective Therapy 53

of culture and susceptibility testing are available, deescala- 7. Levison ME, Levison JH. Pharmacokinetics and pharmacodynamics of
tion should be performed. antibacterial agents. Infect Dis Clin North Am. 2009;23:791-815:vii.
4. Certain combinations of AMD are synergistic for treatment 8. Weese JS. Investigation of antimicrobial use and the impact of anti-
of specific infections. For example, the combination of a microbial use guidelines in a small animal veterinary teaching hos-
pital: 1995-2004. J Am Vet Med Assoc. 2006;228:553-558.
β-lactam and an aminoglycoside is synergistic for treatment
9. Bakken JS. The fluoroquinolones: how long will their utility last?
of enterococcal endocarditis. The relatively slow bactericidal Scand J Infect Dis. 2004;36:85-92.
activity of the β-lactam is greatly enhanced by the addition 10. Jacobs MR. Optimisation of antimicrobial therapy using phar-
of the aminoglycoside, such that shorter durations of treat- macokinetic and pharmacodynamic parameters. Clin Microbiol
ment or extension of the dosing interval becomes possible Infect. 2001;7:589-596.
without affecting outcome.7 11. Preston SL, Briceland LL. Intrathecal administration of amikacin
5. Some drugs are not effective against anaerobic bacteria (e.g., for treatment of meningitis secondary to cephalosporin-resistant
fluoroquinolones with the exception of pradofloxacin). They Escherichia coli. Ann Pharmacother. 1993;27:870-873.
should be combined with an agent active against anaerobes 12. Johnson RH, Einstein HE. Amphotericin B and coccidioidomyco-
(e.g., clindamycin or metronidazole) when a mixed infection sis. Ann N Y Acad Sci. 2007;1111:434-441.
13. Rosanova MT, Lede R, Capurro H, et al. Assessing fluoroquinolones
with anaerobic and aerobic bacteria is suspected.
as risk factor for musculoskeletal disorders in children: a systematic
6. When a vector-borne pathogen is suspected, doxycycline review and meta-analysis. Arch Argent Pediatr. 2010;108:524-531.
may be administered until the results of further diagnos- 14. Sansone JM, Wilsman NJ, Leiferman EM, et al. The effect of fluo-
tic tests confirm or eliminate this suspicion. In these cases, roquinolone antibiotics on growing cartilage in the lamb model.
patients are frequently administered doxycycline in combi- J Pediatr Orthop. 2009;29:189-195.
nation with another active agent to treat alternative bacterial 15. Mertz D, Koller M, Haller P, et  al. Outcomes of early switching
infections that have not yet been ruled out. from intravenous to oral antibiotics on medical wards. J Antimi-
crob Chemother. 2009;64:188-199.
Monitoring the Response to Treatment 16. Scheinfeld NS, Allan JM. Intravenous-to-oral switch therapy.
2011. http://emedicine.medscape.com/article/237521-overview.
Last accessed May 12, 2012.
The response to antimicrobial treatment is best monitored based
17. Papich MG, Davis JL, Floerchinger AM. Pharmacokinetics, protein
on clinical assessment, which includes follow-up cytologic binding, and tissue distribution of orally administered cefpodoxime
examination or cultures. Therapeutic drug monitoring can be proxetil and cephalexin in dogs. Am J Vet Res. 2010;71:1484-1491.
used to ensure adequate serum drug concentrations when treat- 18. Williamson R, Calderwood SB, Moellering Jr RC, et al. Studies on
ing with aminoglycosides, vancomycin, and azole antifungal the mechanism of intrinsic resistance to beta-lactam antibiotics in
drugs. It should be remembered that in some cases, infections group D streptococci. J Gen Microbiol. 1983;129:813-822.
resolve because of the host immune response alone. 19. Arai K, Hirakata Y, Yano H, et al. Emergence of fluoroquinolone-
resistant Streptococcus pyogenes in Japan by a point mutation lead-
SUGGESTED READINGS ing to a new amino acid substitution. J Antimicrob Chemother.
2011;66:494-498.
Hawkey PM, Jones AM. The changing epidemiology of resistance. 20. Hawkey PM, Jones AM. The changing epidemiology of resistance.
J Antimicrob Chemother. 2009;64(suppl 1):i3-i10. J Antimicrob Chemother. 2009;64(suppl 1):i3-i10.
Levison ME, Levison JH. Pharmacokinetics and pharmacodynamics of 21. Bush K, Jacoby GA. Updated functional classification of beta-­
antibacterial agents. Infect Dis Clin North Am. 2009;23(4):791-815. lactamases. Antimicrob Agents Chemother. 2010;54:969-976.
Weese JS. Investigation of antimicrobial use and the impact of antimi- 22. Jacoby GA, Munoz-Price LS. The new beta-lactamases. N Engl
crobial use guidelines in a small animal veterinary teaching hospital: J Med. 2005;352:380-391.
1995-2004. J Am Vet Med Assoc. 2006;228(4):553-558. 23. Jacoby GA. AmpC beta-lactamases. Clin Microbiol Rev.
2009;22:161-182.
REFERENCES 24. Pages JM, James CE, Winterhalter M. The porin and the permeat-
1. Domagk G. Ein Beitrag zur Chemotherapie der bakteriellen Infek- ing antibiotic: a selective diffusion barrier in gram-negative bacte-
tionen. Deut Med Wochenschr. 1935;61:250-253. ria. Nat Rev Microbiol. 2008;6:893-903.
2. Weese JS, Blondeau JM, Boothe D, et  al. Antimicrobial use 25. Li XZ, Nikaido H. Efflux-mediated drug resistance in bacteria: an
guidelines for treatment of urinary tract disease in dogs and update. Drugs. 2009;69:1555-1623.
cats: antimicrobial guidelines working group of the international 26. Sköld O. Resistance to trimethoprim and sulfonamides. Vet Res.
society for companion animal infectious diseases. Vet Med Int. 2001;32:261-273.
2011:2011:263768. 27. Górski A, Miedzybrodzki R, Borysowski J, et  al. Bacteriophage
3. American Association of Feline Practitioners. Basic guidelines of therapy for the treatment of infections. Curr Opin Invest Drugs.
judicious therapeutic use of antimicrobials in cats. 2001. http://www. 2009;10:766-774.
catvets.com/uploads/PDF/antimicrobials.pdf. Last accessed May 28. O’Flaherty S, Ross RP, Coffey A. Bacteriophage and their lysins
12, 2012. for elimination of infectious bacteria. FEMS Microbiol Rev.
4. American Veterinary Medical Association. American Association 2009;33:801-819.
of Feline Practitioners/American Animal Hospital Association basic 29. Findlay B, Zhanel GG, Schweizer F. Cationic amphiphiles, a
guidelines of judicious therapeutic use of antimicrobials. 2006. new generation of antimicrobials inspired by the natural anti-
(updated April 2009) http://www.avma.org/issues/policy/jtua_ microbial peptide scaffold. Antimicrob Agents Chemother.
aafp_aaha.asp. Last accessed May 12, 2012. 2010;54:4049-4058.
5. Morley PS, Apley MD, Besser TE, et al. Antimicrobial drug use in 30. Bai H, Xue X, Hou Z, et al. Antisense antibiotics: a brief review
veterinary medicine. J Vet Intern Med. 2005;19:617-629. of novel target discovery and delivery. Curr Drug Discov Technol.
6. Canadian Veterinary Medical Association. Guidelines on the 2010;7:76-85.
prudent use of antimicrobial drugs in animals. 2000. http:// 31. Lepper MH, Dowling HF. Treatment of pneumococcic meningitis
canadianveterinarians.net/Documents/Resources/Files/85_ with penicillin compared with penicillin plus Aureomycin; studies
Resources_Prudent-Use-of-Antimicrobial-Drugs-in-Animals.pdf. including observations on an apparent antagonism between penicil-
Last accessed May 12, 2012. lin and Aureomycin. AMA Arch Intern Med. 1951;88:489-494.
CHAPTER 7

Antiviral and Immunomodulatory Drugs


Jane E. Sykes and Mark G. Papich

w KEY POINTS
• M  ost antiviral drugs available for treatment of canine and feline the only antiretroviral drug that has demonstrated efficacy for
viral infections are nucleoside analogues with greatest activity treatment of cats, primarily for FIV infections.
against herpesvirus and retrovirus infections. • Antiviral drugs can act synergistically with immunomodulators.
• Because antiviral drugs also affect the function of host cell • Immunomodulators include microbial products, plant-derived
machinery, they have considerable potential for toxicity. immunomodulators, naturally occurring mammalian proteins,
• Antiviral drugs are widely used in human medicine for treat- glucocorticoids, and synthetic compounds such as pentoxifyl-
ment of herpesvirus, HIV and other viral infections. Much less line and opioids. The effect of many of these drugs on outcome
is known about these medications in dogs and cats and there in dogs and cats with infectious diseases has not been fully
are few diseases of dogs and cats for which efficacy has been evaluated using large, prospective, randomized, masked, con-
demonstrated. trolled clinical trials.
• Because of important differences between human and ani- • The parenteral use of natural or recombinant human protein
mal virus infections, one should never assume that an antiviral immunomodulators in dogs and cats may result in the forma-
agent used in people should be used in animals unless there is tion of neutralizing antibodies after 1 to 2 weeks of treatment,
proof of safety and efficacy. which can cross-react with endogenous proteins.
• Antiviral drugs used to treat feline herpesviral infections
include famciclovir, idoxuridine, and cidofovir. Zidovudine is

INTRODUCTION Although all antiviral drugs used to treat small animals were
originally developed for treatment of human viral infections, the
The purpose of this chapter is to describe the mechanisms of application of these drugs to treat viral infections of small animals
action, spectrum of activity, and adverse effects of the major anti- has not proven straightforward. This relates to differences in the
viral and immunomodulatory drugs that have exhibited utility or composition of human and companion animal viruses (which
have potential for treatment of companion animal infectious dis- affect their susceptibility to inactivation by antiviral drugs), or
eases. An enormous number of different compounds have been problems relating to toxicity of human antiviral drugs in dogs
considered or evaluated in vitro and in vivo with variable success. and cats.
Only those currently in use for treatment of dogs and cats with Antiviral drugs can be classified based on their spectrum of
naturally occurring infections, those that show considerable prom- activity (i.e., family of viruses that are inhibited) and their mode
ise for the future, or those evaluated in controlled clinical trials are of action. Antiviral drugs used in small animal medicine are pri-
discussed in this chapter. marily effective against herpesviruses or retroviruses, and most
are nucleoside analogues. Nucleoside analogues resemble host
Antiviral Drugs nucleosides, which are nitrogenous bases with an attached sugar
molecule used as a building block for the formation of DNA
Antiviral agents interfere with virus-specific events in replica- or RNA (Figure 7-1). Use of nucleoside analogues by viruses
tion, including viral attachment, uncoating, assembly, and during replication leads to the formation of abnormal nucleic
virus-directed macromolecular synthesis. As a result, antiviral acids or termination of nucleic acid synthesis. Latent viral infec-
agents typically have a restricted spectrum of activity. In con- tions are not affected because the replication of latent virus is
trast to most antibacterial drugs, antiviral drugs can also affect suspended. However, reactivation of these infections can be
the function of host cell machinery and thus antiviral drugs have reduced in frequency or prevented. Examples of nucleoside ana-
a greater risk of toxicity. In addition to acute toxicities such as logues and their spectrum of activity are shown in Figure 7-1
marrow suppression, many antiviral agents are immunosuppres- and Table 7-1. Other antiviral drugs that have been used in
sive, carcinogenic, and teratogenic. Many are given topically in clinical veterinary medicine include amino acids (l-lysine) and
order to minimize systemic toxicity. Antiviral agents are more neuraminidase inhibitors (oseltamivir). The value of other treat-
likely to be effective when an intact host immune response is ments such as small inhibitory RNA molecules for viral infec-
present. Combinations of antiviral drugs with different mecha- tions of small animals is also under investigation.1
nisms of action and combinations of antiviral and immunomod-
ulatory drugs are increasingly being used to treat human viral Antiherpesviral Drugs
infections, especially HIV infection, with an associated decrease The standard antiviral treatments for herpesviral infections
in toxicity and reduction in selection for drug-resistant mutants. in human patients are the nucleoside analogues acyclovir and

54
CHAPTER 7  Antiviral and Immunomodulatory Drugs 55

penciclovir, and their prodrugs valacyclovir and famciclo- DNA polymerases of different herpesviruses are inhibited to
vir, respectively. These drugs are activated by the herpesviral varying degrees by acyclovir triphosphate, not all herpesviral
enzyme thymidine kinase (TK), which phosphorylates them to infections are equally susceptible. Drug resistance results from
a monophosphate form. Host cell enzymes then phosphorylate reduced viral TK activity, altered viral TK, or altered viral DNA
the drugs further to triphosphate forms, which concentrate in polymerase. The antiherpesviral drugs cidofovir, idoxuridine,
virus-infected cells and interfere with viral DNA replication via trifluridine, and vidarabine are not dependent on viral TK for
inhibition of the viral DNA polymerase enzyme. Because the phosphorylation and so have greater host cell toxicity. In small
animal medicine, these four drugs have been used topically to
treat ocular feline herpesviral infections.

Acyclovir and Valacyclovir


Acyclovir is a synthetic analogue of the purine nucleoside deoxy-
guanosine (see Figure 7-1). In human patients, acyclovir is widely
used to treat herpes simplex and varicella-zoster virus infections,
both of which are α-herpesviruses. In small animal medicine,
acyclovir has primarily been used to treat feline herpesvirus-1
(FHV-1) infections, although FHV-1 has a much lower suscepti-
bility to the drug compared with the human herpesviruses.2 Acy-
A C clovir has also been used to treat canine herpesvirus-1 (CHV-1)
infections in puppies (see Chapter 16). Other nucleoside ana-
logues (trifluridine, idoxuridine, cidofovir, ganciclovir, and
penciclovir) show much greater activity than acyclovir against
FHV-1 in vitro.2-4
Topical acyclovir has been used with limited success to treat
feline herpetic dermatitis5 and keratitis.6 Topical treatment
requires frequent (>4 times a day) application for beneficial
effect.6 The oral bioavailability of acyclovir is low in cats, and
high doses are required to achieve adequate serum drug con-
centrations.7 Valacyclovir, the prodrug for acyclovir, is rapidly
converted to acyclovir by a first-pass effect after oral adminis-
B D tration, and administration of the prodrug results in improved
FIGURE 7-1  Chemical structures of deoxyguanosine (a nucleoside) and antiherpes- oral bioavailability of acyclovir. Unfortunately, administration
viral nucleoside analogues. the grey portion is missing in the nucleoside analogues. A, of high doses of acyclovir or valacyclovir to cats has resulted in
Deoxyguanosine. B, Acyclovir. C, Penciclovir. D, Ganciclovir. significant toxicity, including myelosuppression, renal tubular

TABLE 7-1
Antiviral Drugs in Use in Small Animals

Antiviral Drug Mechanism of Action Human Applications Small Animal Applications


Acyclovir/­ Guanosine analogue; interferes with Herpesviruses, especially HSV Poorly effective against FHV-1
valacyclovir viral DNA polymerase and DNA and varicella-zoster virus
synthesis. Activity requires viral TK.
Penciclovir/­ See acyclovir Herpesviruses, especially HSV FHV-1 infections
famciclovir and varicella-zoster virus
Cidofovir Deoxycytidine monophosphate Systemically to treat cytomegalo- Topical treatment of FHV-1
­analogue. Activity independent of virus ­retinitis; topically to treat ocular infections
viral TK. ­papillomavirus infections
Idoxuridine Iodinated thymidine analogue. Topical treatment of HSV kerato- Topical treatment of FHV-1
­Interferes with viral DNA ­synthesis. conjunctivitis keratitis
Trifluridine Fluorinated thymidine analogue. See idoxuridine See idoxuridine
­Interferes with viral DNA ­synthesis.
Vidarabine Adenosine analogue. Interferes with See idoxuridine See idoxuridine
viral DNA synthesis.
Zidovudine Thymidine analogue. Interferes with Systemic treatment of HIV Systemic treatment of FIV and
viral DNA synthesis. ­infections FeLV infections

FHV-1, feline herpesvirus-1; HSV, herpes simplex virus; TK, thymidine kinase.
56 SECTION 2  Antiinfective Therapy

necrosis, and hepatic necrosis, without effective suppression ganciclovir holds promise for treatment of feline ocular herpes-
of viral replication.8 Thus the use of systemic acyclovir and viral infections.
valacyclovir to treat cats with herpesvirus infections is not
recommended. Cidofovir
Cidofovir is a nucleotide analogue of deoxycytidine mono-
Penciclovir and Famciclovir phosphate. Because the drug already has a monophosphate
Of all antiviral drugs used to treat small animal patients, group, its metabolism to the active diphosphate form by host
­penciclovir and famciclovir have shown the greatest promise. cellular enzymes is not dependent on viral TK, and so it has
Penciclovir is another guanosine analogue (see Figure 7-1). It been used to treat acyclovir- and penciclovir-resistant infec-
is present at much higher concentrations and for longer dura- tions. In human patients, cidofovir is primarily used to treat
tion in cells than is acyclovir, which permits less frequent dos- cytomegalovirus retinitis. Cidofovir also has efficacy against
ing. In humans, the prodrug famciclovir is well absorbed orally other DNA virus infections, such as poxvirus and papillomavi-
and rapidly converted to penciclovir, leading to increased oral rus infections, and has been used topically as a cream to treat
bioavailability of penciclovir. Most of the drug is eliminated viral warts in people. Because the oral bioavailability of cido-
unchanged in the urine, and so dose reduction may be required fovir is extremely low (<5%), it is always administered intrave-
for animals with decreased kidney function. In human patients, nously, intravitreally, or topically. Cidofovir has a prolonged
the concurrent administration of food decreases peak plasma intracellular half-life, which enables infrequent parenteral dos-
concentrations without affecting overall bioavailability. In ing regimens in human patients (once weekly, and then every
humans with herpes simplex virus and herpes zoster infections, other week for maintenance therapy).
famciclovir is as effective as acyclovir. In contrast, penciclovir Like ganciclovir, cidofovir is highly active in  vitro against
(and thus famciclovir) have been shown to be potent inhibi- FHV-1.4,14,15 Twice-daily administration of a 0.5% cidofo-
tors of FHV-1 replication (which is not the case for ­acyclovir). vir ophthalmic solution significantly decreases viral shedding
­Penciclovir-resistant mutants of FHV-1 with altered TK enzymes and the severity of clinical disease in cats with experimentally
have been described.9 induced ocular FHV-1 infection (Table 7-2).16 Local irritation
Administration of famciclovir to cats at dosages comparable and scarring of the nasolacrimal duct has been reported with
to those used in other species results in much lower plasma con- topical administration of cidofovir to humans and rabbits with
centrations and a longer time to development of peak plasma keratoconjunctivitis.
concentrations of penciclovir when compared with other ani-
mal species, which suggests altered absorption or metabolism Idoxuridine and Trifluridine
of famciclovir in cats.10 High doses of famciclovir have been Idoxuridine and trifluridine are halogenated thymidine ana-
required to achieve adequate plasma drug concentrations. Nev- logues that interfere with the replication of FHV-1 in  vitro,
ertheless, famciclovir is well tolerated when administered orally although they are more potent inhibitors of herpes simplex virus
to cats at doses that produce clinical responses.5,10,11 Treat- replication.2,14 They have been used to treat herpesviral kerati-
ment of cats with experimentally induced FHV-1 conjunctivitis tis in humans and in cats.17 These drugs are highly toxic when
with famciclovir at 90 mg/kg PO q8h for 21 days resulted in given systemically, because host cell and viral DNA synthesis are
lower clinical and pathologic disease scores and decreased viral equally affected. Frequent topical application is required (five to
shedding when compared with placebo-treated cats.11 Clinical six times daily), and prolonged use can cause corneal irritation
responses also occur in naturally infected cats with both acute or ulceration. Trifluridine has better corneal penetration than
and chronic manifestations of disease with negligible adverse idoxuridine and is available as a 1% ophthalmic solution. Unfor-
effects.5 Because of saturation of the metabolism of famciclovir tunately, trifluridine is expensive, and ocular administration of
to penciclovir, equivalent serum and tear penciclovir concentra- trifluridine is often extremely irritating to cats.18 In contrast,
tions can be achieved in cats with 40 or 90 mg/kg PO q8h of topical idoxuridine administration is generally well tolerated.
famciclovir, so 40 mg/kg PO q8h is considered equally effica-
cious.12,13 Clinical improvement often occurs within a week of Vidarabine
treatment. Vidarabine is an adenosine analogue that is phosphorylated by
host cellular enzymes to vidarabine triphosphate, which inter-
Ganciclovir feres with DNA synthesis by both the virus and host cells. It
Ganciclovir resembles acyclovir except that it has an additional is effective against idoxuridine-resistant herpesviral strains
hydroxymethyl group on its acyclic side chain (see Figure 7-1). because its mechanism of action differs from that of idoxuri-
In human patients, ganciclovir is widely used specifically for dine. It is reportedly well tolerated by cats when administered
the treatment of human cytomegalovirus infections, which can five to six times daily to cats as a 3% ophthalmic ointment.18
be life threatening in the immunocompromised. Cytomegalo-
viruses are β-herpesviruses. Systemic administration of ganci- Lysine
clovir to human patients is associated with a high prevalence Lysine is an amino acid that interferes with herpesviral repli-
of adverse drug reactions, especially cytopenias and central cation by a poorly understood mechanism. Antagonism of
nervous system (CNS) signs, and so the use of ganciclovir is arginine may somehow be involved, because a high lysine-to-
limited to patients with life-threatening or sight-threatening arginine ratio appears to be important for efficacy. However,
infections. A topical ophthalmic gel formulation of ganciclo- arginine itself was also shown to interfere with the replication
vir (0.15%) is now available for treatment of keratitis caused of herpes simplex virus in vitro.19
by herpes simplex virus-1. Ganciclovir is highly inhibitory to Lysine has shown efficacy when administered as tablets
FHV-1 replication in  vitro.4,14 Pharmacokinetic, safety, and to cats with FHV-1 conjunctivitis20 and, in another study, it
efficacy studies in cats have not yet been performed, but topical reduced shedding of reactivated virus by latently infected cats.21
CHAPTER 7  Antiviral and Immunomodulatory Drugs 57

TABLE 7-2
Antiviral Drugs Used for Treatment of Feline Herpesvirus Infections
Drug Dose Interval Route Comments
Famciclovir 62.5 mg/cat, 125 mg/cat, q8h PO Reduce dose for cats with renal insufficiency.
or 40-90 mg/kg Compound suspensions are bitter and
poorly tolerated. Broken tablets are best
administered in a gel cap. Safe in kittens.
Cidofovir 0.5% solution q12h Topical ophthalmic Monitor for ocular irritation. Solution
prepared from commercial 7.5% intrave-
nous solution by dilution in sterile saline.
Store diluted solution up to 6 months
at 4°C, −20°C, and −80°C. Can also be
obtained from compounding pharmacies.
Idoxuridine 0.1% solution or 0.5% 5-6 times daily Topical ophthalmic Monitor for ocular irritation and ulcer-
ointment ation. Compounding may be required.
Trifluridine 1% solution 5-6 times daily Topical ophthalmic May be poorly tolerated.
Vidarabine 3% ointment 5-6 times daily Topical ophthalmic Well tolerated. Compounding may be
required.
Lysine 250 mg (kittens) q12h PO Questionable efficacy. Bolus administration
500 mg (cats) preferred to dietary supplementation.
Zidovudine 5 to 15 q12h PO, SC Monitor CBC weekly during treatment for
the first month, then monthly. Use low
end of the dose range in renal failure. Use
higher dose with caution.

When administered as tablets to cats in a shelter, there was no HIV. Inside host cells, it is converted to the active triphosphate
reduction in upper respiratory tract disease.22 There was concern form. AZT inhibits the replication of FIV, reduces plasma viral
that the stress of tablet administration may have contributed to load, improves stomatitis, and increases CD4/CD8 ratios in cats
disease in these cats. However, in two other studies, dietary sup- naturally infected with FIV.27,28 Some FIV isolates are resistant as
plementation with lysine was not effective for management of a result of mutations in the RT enzyme. Although AZT is active
upper respiratory tract disease in a shelter.23,24 In fact, cats that against FeLV in vitro, when compared with FIV-infected cats, it
received the lysine-supplemented diet had more severe disease has not performed as well for treatment of naturally infected, sick
and more frequent viral shedding than cats that received a non- cats with chronic FeLV infection. Nevertheless, improvement in
supplemented ration, despite having increased plasma lysine stomatitis, reduced antigenemia, and reduction in development of
concentrations. The lysine-supplemented diet did not affect lymphoma have been reported in studies of naturally and experi-
plasma arginine concentration, so altered arginine levels did not mentally FeLV-infected cats that were treated with AZT.28,29
appear to contribute to the increased severity of disease. AZT is available as a10 mg/mL syrup and a 10 mg/mL injec-
tion. It has good oral bioavailability and is well distributed to
Antiretroviral Drugs tissues, including the CNS. It is metabolized to an inactive form
All antiviral drugs used to treat cats with retrovirus infections by the liver and excreted by the kidneys. Dosage reduction has
have been nucleoside analogues, which inhibit the DNA poly- been recommended for cats with renal failure. Unfortunately,
merase function of the retroviral reverse transcriptase (RT) some cats treated with AZT can develop dose-related hemato-
enzyme. Unfortunately, many drugs used for treatment of HIV logic adverse effects, most commonly nonregenerative anemia
infections (such as protease inhibitors) are not effective for and neutropenia, so the CBC must be monitored during treat-
treatment of feline retrovirus infections, because they only act ment. Adverse effects may be confused with retrovirus-induced
on HIV enzymes. The only antiretroviral that has shown benefit cytopenias. In human patients with HIV infection, cytopenias
in naturally infected cats is zidovudine (AZT). At the time of are more likely to occur when disease is advanced.
writing, an integrase inhibitor known as raltegravir has shown Fozivudine is a thioether lipid-zidovudine conjugate that
early promise for treatment of FeLV infections both in vitro and undergoes intracellular cleavage to zidovudine monophos-
in vivo.25,26 Many drugs that have activity against feline retrovi- phate and subsequent phosphorylation to the active triphos-
ruses in vitro, such as ribavirin and adefovir (PMEA), are toxic phate form. Cleavage preferentially occurs in lymphocytes and
when given to cats, which limits their use in practice. monocytes compared with RBC and marrow stem cells, and so
hematologic toxicity is less likely to occur. Fozivudine reduces
Zidovudine and Fozivudine viremia in cats experimentally infected with FIV, without signif-
Zidovudine (azidothymidine; AZT; Retrovir) is a thymidine ana- icant adverse effects.30 Further study is required to evaluate this
logue and was one of the first drugs shown to be effective against drug for treatment of chronic FIV and FeLV infections in cats.
58 SECTION 2  Antiinfective Therapy

Lamivudine
Lamivudine (3TC), a cytidine analogue, is synergistic when
combined with AZT for treatment of HIV infection, and the
combination is in common use in human medicine. AZT/3TC
prevented FIV infection when given to cats shortly after experi-
mental inoculation, but did not appear to be beneficial for treat-
ment of cats with chronic FIV infection.31 In addition, severe
hematologic adverse effects and fever occurred in some cats.

Other Antiretroviral Drugs


Raltegravir inhibits the retroviral integrase enzyme. Integrase
incorporates transcribed viral DNA into the host chromosome.
In human patients, raltegravir is approved for treatment of HIV
infections and is used in combination with other antiretroviral
drugs. Although expensive, raltegravir has shown great promise
for treatment of FeLV infections in vitro and also in vivo.25,26
Other drugs that have shown promise in vitro for treatment of
FeLV infections, with no evidence of toxicity to cell cultures,
are the nucleoside analogues tenofovir, decitabine, and gem-
citabine.26 Tenofovir is used as part of combination therapy to FIGURE 7-2  Structure of an influenza virus. The M2 protein, which is only ­present in
influenza A viruses, is inhibited by tricyclic amines such as amantadine. ­Oseltamivir inhib-
treat HIV infections. It is administered as a prodrug. Decitabine
its the viral neuraminidase.
and gemcitabine are cytidine analogues used in human patients
to treat myelodysplastic syndromes and carcinomas, respec-
tively. The use of combinations of gemcitabine and carboplatin excretion. The drug is well tolerated in humans, with gastroin-
to treat carcinomas in cats has been reported, but cytopenias testinal signs being the most frequently reported adverse effects.
and gastrointestinal toxicity occurred in some of the cats.32 The prevalence of resistance to oseltamivir among influ-
Plerixafor is a bicyclam derivative that selectively blocks the enza virus isolates is generally low (<5%). High-level resistance
chemokine receptor, CXCR4. This receptor is used by FIV to results from mutations in the viral neuraminidase. Neuramini-
enter cells (see Chapter 21). In a placebo-controlled, masked dase inhibitors could play a critical role in prevention of mor-
clinical trial, administration of plerixafor to cats with FIV tality in human influenza virus pandemics, so strategies that
infection for 6 weeks reduced proviral load but did not lead to minimize the selection of resistant mutants are important. The
improvement in clinical or immunologic variables.33 Centers for Disease Control and Prevention recommends priori-
tizing antiviral treatment to human patients at risk of complica-
Antiinfluenza Viral Drugs tions of influenza, such as the very young and very old.36
The main drugs used in human medicine to treat influenza virus Oseltamivir has been used to treat canine parvovirus (CPV)
infections are neuraminidase inhibitors, such as oseltamivir and enteritis in puppies, with anecdotal reports of improved ­outcome.
zanamivir, and the tricyclic amines amantadine and rimanta- A single prospective, randomized, masked, ­placebo-controlled
dine, which inhibit the M2 ion channel protein that is present in trial of 35 dogs with CPV enteritis showed that dogs treated with
influenza A viruses (Figure 7-2). Only oseltamivir has been used oseltamivir (2 mg/kg PO q12h) had no significant drop in their
to any great extent in small animals. Amantadine, which also white blood cell count, whereas untreated dogs had a significant
has antagonistic effects at the NMDA (N-methyl-d-aspartate) drop in their white blood cell count in the first 5 days of hospi-
receptor, has been used to treat osteoarthritis in animals in com- talization.37 Treated dogs also gained weight during hospitaliza-
bination with other drugs.34 tion, whereas untreated dogs lost weight. However, there was
no difference in hospitalization time, treatments needed, clinical
Oseltamivir scores, morbidity, or mortality between the two groups, and the
Oseltamivir (Tamiflu) is the prodrug of oseltamivir carboxylate number of dogs in each group was small. No significant adverse
(GS4071), a potent inhibitor of influenza virus neuraminidase. drug effects were observed, but oseltamivir was ­administered
Oseltamivir was developed because of the poor oral bioavailabil- as a 1:1 dilution with water, in order to reduce reactions to the
ity of zanamivir, a neuraminidase inhibitor that is structurally taste of the drug and vomiting shortly after drug administration.
similar to GS4071. Neuraminidase is a surface glycoprotein of The authors acknowledged that there were potential concerns
both influenza A and influenza B viruses (see Figure 7-2). The that related to administration of an oral medication to dogs
viral neuraminidase cleaves sialic acid residues on the surface of with enteritis, with variability in drug absorption.
infected cells, which allows new virus particles to be released from As CPV has no neuraminidase, it was hypothesized that osel-
host cells. It also prevents aggregation of virus particles after they tamivir instead may act on the neuraminidases of bacteria that
are released and facilitates spread of the virus through the mucus are normally responsible for secondary bacterial infections in
of the respiratory tract by cleaving sialic acid residues in mucin. CPV enteritis, which are primarily those of the gastrointesti-
In humans and dogs, oseltamivir has high oral bioavailabil- nal tract. The role of bacterial neuraminidases in the pathogen-
ity.35 Esterase enzymes in the liver then convert oseltamivir to esis of enteric bacterial infections and bacterial translocation is
its active form, which is well distributed to most body fluids, unknown. Bacterial neuraminidase enzymes may play a role in
including surface epithelial cells throughout the upper and biofilm formation and help bacteria to invade mucin layers of
lower respiratory tract. Elimination of the drug relies on renal the respiratory tract.38 It has been suggested that gut bacteria
CHAPTER 7  Antiviral and Immunomodulatory Drugs 59

may use neuraminidase enzymes to cleave sialic acid residues on result in cure. The use of human recombinant cytokines beyond
gastrointestinal epithelial cells, which exposes receptor sites for 1 or 2 weeks of treatment has been associated with the develop-
bacterial adherence. ment of antibodies against the cytokines, which have the poten-
A major concern that relates to treatment of CPV enteri- tial to cross-react with endogenous cytokines. In the absence
tis with oseltamivir is the possibility of selection for resistant of data from randomized, placebo-controlled clinical trials to
mutants among influenza viruses if widespread use of the drug support their use, immunomodulators should be used with cau-
occurs in veterinary clinics. Given the restrictions on the use of tion, because they may have the potential to accelerate disease.
this drug for treatment of human influenza virus infections, fur- This may occur for viruses that are dependent on rapidly divid-
ther investigation is required before the use of oseltamivir can ing cells for replication (such as parvovirus) or those that rep-
be recommended for treatment of CPV enteritis. licate only in activated lymphocytes (such as FIV). Promotion
of an inflammatory response may also have detrimental effects
Other Antiviral Drugs if immune-mediated processes play an important role in the
Although not useful for treatment of cats because of toxicity, pathogenesis of disease (such as for FIP).
the nucleoside analogue ribavirin has shown promise in  vitro
for treatment of canine distemper virus (CDV) infections. Microbial Products
Another drug, 5-ethynyl-1-beta-d-ribofuranosylimidazole-4- Microbial products with immunomodulatory activity include
carboxamide (EICAR), has also shown activity against CDV bacillus Calmette-Guérin extract (a mycobacterial cell wall
in  vitro39 and for treatment of measles virus, which is closely extract from Mycobacterium bovis), purified staphylococcal
related to CDV.40 Ribavirin is primarily used to treat hepatitis C protein A, killed Propionibacterium acnes (ImmunoRegulin,
virus infections in humans. The pharmacokinetics and toxicity ImmunoVet), Staphylococcus aureus phage lysate (Staph-
of ribavirin and EICAR in dogs require further study. age Lysate, Delmont Laboratories, USA), Serratia marcescens
Nelfinavir is a protease inhibitor used to treat HIV infec- abstract, and inactivated poxviruses (Baypamun, Bayer). To
tion. It also limits the replication of severe acute respiratory date, controlled clinical trials with these products have not dem-
syndrome (SARS) coronavirus in vitro,41 but its mode of action onstrated significant efficacy for treatment of viral infections
against coronaviruses is poorly understood. Nelfinavir was in dogs and cats. Staphylococcal phage lysate, which contains
not effective in a mouse model of SARS.42 Although nelfinavir components of S. aureus and a bacteriophage, and Propionibac-
alone was not completely effective, a combination of nelfinavir terium acnes immunotherapy have shown efficacy for treatment
and Galanthus nivalis agglutinin (GNA) inhibited the replica- of pyoderma in dogs and are available commercially for this
tion of feline coronavirus in  vitro and may have application purpose (see Table 7-3; also see Chapter 84).45
for the treatment of feline infectious peritonitis (FIP).43 GNA
appears to bind the viral outer envelope glycoproteins and Plant-Derived Biologic Response Modifiers
prevent cellular entry. The use of nelfinavir was reported in a Acemannan (Carrisyn, Carrington Laboratories, Irving, TX) is
few cats with naturally occurring coronavirus infections,43,44 a mucopolysaccharide derived from aloe vera plant leaves that
but the toxicity and pharmacokinetics of nelfinavir and GMA induces cytokine production and dendritic cell maturation. It
in cats and efficacy of these drugs for treatment of FIP remains stimulates macrophages to secrete a variety of cytokines, includ-
unknown. ing IFN-γ, TNF-α, prostaglandin E2, Il-1, and Il-6. It has been
used to treat FIV and FeLV infections, either by oral or paren-
Immunomodulators teral administration,46,47 and a veterinary product is available
for treatment of canine and feline fibrosarcomas. Controlled
An immunomodulator is any drug that alters immune system trials are needed to understand the efficacy of this treatment in
function. Immunomodulators are also referred to as biologic naturally infected cats.
response modifiers. Their use for treatment of infectious dis- Polyprenyl immunostimulant (Sass & Sass, Inc., Oakridge,
eases may be beneficial when compromise of the immune system TN) is a plant-derived investigational veterinary biological that
impairs effective antimicrobial drug treatment. Alternatively, is made up of phosphorylated, linear polyisoprenols. In vitro, it
immunomodulators can be used to dampen an excessive host upregulates synthesis of Th1 cytokines and has antiviral proper-
inflammatory response. ties. Cats with experimentally induced FHV-1 infection had a
A variety of immunomodulators have been used to treat decreased severity and duration of upper respiratory tract signs
feline retroviral infections, FIP, FHV-1 infections, and canine when treated with polyprenyl immunostimulant, in compari-
and feline parvoviral enteritis (Table 7-3). Immunomodulators son to placebo-treated cats.47a Long-term (>2 years) survival
used in veterinary medicine can be divided into six main groups: has been reported in a few cats with noneffusive FIP that were
(1) microbial products; (2) plant-derived biologic response treated with polyprenyl immunostimulant. The response in cats
modifiers; (3) naturally occurring cytokines, such as colony- with effusive FIP has been disappointing.48
stimulating factors, and interferons; (4) immunoglobulins;
(5) glucocorticoids; and (6) synthetic compounds with immuno- Naturally Occurring Mammalian Proteins
modulatory activity, such as pentoxifylline. Colony-Stimulating Factors
Treatment of canine and feline viral infections with immuno- Colony-stimulating factors (CSFs) are naturally occurring gly-
modulators has frequently been associated with disappointing coproteins that stimulate production, differentiation, survival,
results, and data from large, prospective, controlled clinical tri- and activation of white blood cells. Examples include erythro-
als are generally not available. Frequently, an immunomodula- poietin; thrombopoietin; Il-5 (which stimulates eosinophil and
tor shows antiviral activity in vitro but this does not correlate basophil production); Il-3; stem cell factor; and macrophage
with treatment responses in  vivo. Sometimes, mild to moder- CSF, granulocyte CSF (G-CSF), and granulocyte-macrophage
ate clinical improvement is observed, but treatment does not CSF (GM-CSF). Recombinant G-CSF has been used most
60 SECTION 2  Antiinfective Therapy

TABLE 7-3
Commercially Available Immunomodulators That May Have Benefit for Treatment of Infectious Diseases of Dogs and Cats
Drug Dose Interval Species Route Comments
Acemannan 2 mg/kg Weekly C SC For treatment of fibrosarcomas,
efficacy unclear for FIV and
FeLV infections
Feline recombinant 1 million U/kg q24h for 5 consecutive C, D SC For FeLV infections
interferon omega days on days 0, 14,
and 60
0.1 million U/kg q24 C Oromucosal For refractory caudal stomatitis
associated with FCV infection
Filgrastim 5 µg/kg q24h C, D SC Monitor CBC weekly during
treatment. Long-term use
(>2 weeks) can lead to
development of neutralizing
antibodies.
Lactoferrin 200 mg powder q24h C Topical For refractory gingivitis and
or 40 mg/kg stomatitis. Must be purchased
solution from chemical suppliers
Lymphocyte T-cell 1 mL Weekly C SC For treatment of FeLV and FIV
­immunomodulator infections
Human recombinant 104 to 106 U/kg q24h C SC Monitor CBC weekly during
interferon alpha 1 to 50 U/cat q24h PO parenteral treatment. Long-
term parenteral use can lead
to development of neutralizing
antibodies
Pentoxifylline 15 mg/kg q8h D PO Benefits for treatment of FIP
¼ of a 400 mg q8h C unclear. Co-administration
tablet of cytochrome P450 inhibi-
tors may increase serum drug
concentrations
Polyprenyl 3 mg/kg Three times weekly C PO Possible benefit for treatment of
­immunostimulant noneffusive FIP
Prednisolone 1 to 2 mg/kg q24h C PO For FIP
Propionibacterium 0.1 mg dogs <7 kg Twice weekly. After D IV For pyoderma. Available as a
acnes, killed 0.2 mg 7-20 kg 14 d, weekly ­until 0.4 mg/mL solution
­remission, and
0.4 mg 20-34 kg
monthly thereafter to
0.8 mg > 34 kg maintain remission
Staphylococcus 0.5 mL Once or twice weekly D SC For pyoderma
­aureus phage lysate

C, Cats; D, dogs.

widely in veterinary medicine as an immunomodulator. The use myelosuppression, for treatment of severe chronic neutropenia,
of these drugs may be limited by their cost and immunogenicity. for mobilization and collection of peripheral blood stem cells
Granulocyte colony-stimulating factor.  Granulocyte CSF for bone marrow transplantation, and to accelerate myeloid
stimulates growth and differentiation of the neutrophil cell line. reconstitution after hematopoietic stem cell transplantation.49
It also stimulates neutrophil function, including chemotaxis, G-CSF and GM-CSF have been used to treat pneumonia and
phagocytosis, antibody-dependent cellular cytotoxicity, and severe sepsis in humans without neutropenia, but significant
superoxide production. Nonglycosylated, recombinant human benefit has not been identified.50,51
G-CSF is available under the trade name Neupogen. Pegfil- When G-CSF is administered as a daily subcutaneous injec-
grastim (Neulasta) is a pegylated form with a long half-life and tion to dogs and cats, the neutrophil count increases over sev-
single-dose administration. Lenograstim is a glycosylated form eral days. This results from an enhanced rate of granulopoiesis
available outside the United States. These three forms have simi- and a shortened neutrophil maturation time. Slight increases
lar efficacy and adverse effects in humans. G-CSF is approved in monocyte and lymphocyte counts can also occur.52 Counts
for use in humans for treatment of chemotherapy-induced return to normal 5 days after discontinuation of treatment.
CHAPTER 7  Antiviral and Immunomodulatory Drugs 61

In veterinary medicine, filgrastim has been used to treat che- of recombinant human and natural IFN-α preparations are avail-
motherapy-induced neutropenia and cyclic neutropenia in gray able, including pegylated forms that have prolonged biologic
collies. Filgrastim was not effective for treatment of leukopenia half-lives. In human patients, IFN-α is used in conjunction with
in experimentally induced CPV infection.53,54 Repeated admin- antiviral drugs to treat hepatitis B and hepatitis C virus infec-
istration of filgrastim to dogs and cats has been associated with tions, papillomavirus infections, and HIV-associated Kaposi’s
development of antibodies to the protein within 2 to 3 weeks of sarcoma. It is generally administered for 24 or 48 weeks.
treatment, which can cross-react with endogenous G-CSF. Thus, Recombinant human IFN-α (rhIFN-α) has been administered
although it can increase neutrophil counts in some FIV-infected parenterally and orally to cats for treatment of feline retroviral
cats, long-term use of filgrastim is not recommended.55 Recom- infections, feline upper respiratory viral infections, and FIP. It
binant canine G-CSF (rcG-CSF) may be safer for long-term use inhibits replication of FeLV, FIV, FHV-1, and feline coronavirus
in dogs, and possibly also cats. One recombinant canine formu- in vitro. Unfortunately, mixed results have been obtained after par-
lation mobilized neutrophils in dogs within 24 hours of admin- enteral and oral administration to cats with retrovirus infections,
istration.56 Recombinant canine G-CSF reduces the severity and and parenteral IFN-α was not effective for treatment of FIP.61 Par-
duration of bone marrow suppression induced by cyclophos- enteral administration can lead to the development of neutralizing
phamide in dogs.57 In a prospective, non-randomized clinical antibodies and apparent loss of activity after 3 or 7 weeks.62
trial for treatment of CPV infection, neutrophil counts were Oral administration of a low dose of rhIFN-α to treat viral
higher and hospital times were shorter in dogs treated with rcG- infections of animals has been controversial because of proteo-
CSF, but survival times were decreased.58 Treatment of canine lytic degradation of rhIFN-α by gastric acid. Nevertheless, ben-
and feline parvovirus infections with G-CSF might cause harm, eficial outcomes have been reported in some cats with FHV-1,
because the increased cell turnover induced by the drug might FeLV, and FIV infections, presumably because immunomodula-
promote parvovirus replication. Further studies are required to tion follows mucosal absorption of the drug. Topical application
evaluate the utility of rcG-CSF for treatment of neutropenias of rhIFN-α to the eye has been used to treat FHV-1 keratitis, but
associated with canine parvoviral enteritis, feline retrovirus controlled studies using this treatment are lacking. With the avail-
infections, and chronic canine monocytic ehrlichiosis. ability of more effective antiviral drugs such as famciclovir, it is
In dogs and cats, exogenous G-CSF is well tolerated. In likely that the use of these antiviral drugs may take precedence
human patients, the main adverse effects of treatment are bone over, or be used in combination with, treatment with IFN-α.
and musculoskeletal pain, headache, anemia, thrombocytope- Feline recombinant interferon-omega.  IFN-ω is a Type 1 inter-
nia, and splenomegaly. Long-term use in humans can be associ- feron closely related to IFN-α. It is secreted by virus-infected
ated with osteopenia as a result of upregulation of osteoclast leukocytes. Its precise mechanism of action is not known. In
activity. Rare complications in people include splenic rupture dogs it increases macrophage and NK cell activity, and it has
and exacerbation of autoimmune disorders. antiviral activity against several feline viruses in vitro, including
Granulocyte-macrophage colony-stimulating factor.  GM-CSF FeLV, FHV-1, feline calicivirus (FCV), canine and feline par-
stimulates growth and function of neutrophils, monocytes, and voviruses, and feline coronavirus. It also has antitumor activ-
eosinophils. Commercial GM-CSF formulations available for ity.63 A recombinant feline formulation of IFN-ω produced in
treatment of hematologic disorders in human patients include silkworms is available in Europe and Canada for veterinary use
sargramostim, molgramostim, and regramostim, which vary (rfIFN-ω, Virbagen omega, Virbac). Because it has similar activ-
slightly in amino acid sequence and degree of glycosylation. ity in canine and feline cells, it has potential for treatment of
When used to treat cats with FIV, viral load increased,59 and both canine and feline viral infections. The optimal dosing regi-
so the use of GM-CSF is not recommended for treatment of men is unknown, but the manufacturer recommends three cycles
FIV-induced cytopenias. Interestingly, sargramostim has been of treatment, on days 0, 14, and 60, for 5 consecutive days.
used to treat some human patients with AIDS-associated oppor- Preliminary data suggested that treatment of cats with
tunistic infections, which include oropharyngeal candidiasis FIP with rfIFN-ω might increase survival times,64 but a
and disseminated mycobacterial infections. Administration of ­randomized, placebo-controlled study of naturally infected
recombinant human GM-CSF to dogs was associated with anti- cats demonstrated no improvement in quality of life or sur-
body development after 10 days.60 vival time.65 When cats with naturally occurring FeLV-related
disease were treated with rfIFN-ω in a placebo-controlled trial,
Interferons survival time over a 2-month follow-up period increased.66
Interferons are cytokines with antiviral properties. IFN-α, IFN- Improvement in clinical scores and laboratory parameters
β, and IFN-ω are type I interferons, which are produced by leu- was also documented in some sick cats that were naturally
kocytes and fibroblasts in response to a viral infection. Type I infected with FeLV and FIV, despite no significant changes in
interferons activate natural killer (NK) cells, increase expression viral load.67 The drug also has been used topically, orally, and
of major histocompatibility complex (MHC) class I molecules, parenterally to treat FHV-1 infections, and subcutaneously to
and have antitumor activity. IFN-γ is the only type II interferon treat refractory FCV-associated stomatitis.68-71 Oromucosal
and is produced by T lymphocytes and NK cells in response to administration of rfIFN-ω to cats with refractory FCV-asso-
antigenic stimulation. IFN-γ plays a critical role in the clear- ciated stomatitis led to significant clinical improvement when
ance of intracellular pathogens by macrophages. Recombinant compared with placebo.68 Treatment of puppies with natu-
human IFN-γ has been used to treat chronic granulomatous rally and experimentally induced CPV enteritis with rfIFN-ω
disease in humans as well as chronic infections caused by intra- in a number of placebo-controlled trials was associated with
cellular bacteria and fungi, but recombinant veterinary prepara- reduced disease severity and, in some studies, significantly
tions are not available. reduced mortality. In contrast, there was no improvement in
Interferon alpha.  Naturally occurring IFN-α represents a survival in cats from a cattery with feline panleukopenia virus
group of more than 20 molecules that vary slightly in composi- infection that were treated with rfIFN-ω when compared with
tion and inhibit viral nucleic acid and protein synthesis. A variety an untreated control group.72
62 SECTION 2  Antiinfective Therapy

Recombinant fIFN-ω is well tolerated by dogs and cats.


Transient lethargy, fever, vomiting, mild diarrhea, and anorexia
have been documented in some treated cats, especially at higher
doses (2.5 × 106 U/kg). Mild neutropenia, eosinophilia, and
reversible increases in the activity of serum AST have also been
described following treatment.73 Additional studies of the effi-
cacy and safety of rfIFN-ω in larger numbers of sick cats with
retroviral infections and cats and dogs with other viral infec-
tions are required.

Lymphocyte T-cell Immunomodulator


Lymphocyte T-cell immunomodulator (T-cyte Therapeu-
tics, Inc.) is a protein derived from thymus epithelial cells.
According to the product brochure, LTCI upregulates Il-2
production in vitro and regulates CD4 T-cell function. It was
also reported to stimulate platelet production in mice with
­chemotherapy-induced thrombocytopenia. LTCI has been
conditionally approved by the U.S. Department of Agriculture
as an aid for treatment of FIV and FeLV infections, and for the
associated signs of cytopenias and opportunistic infections.
Controlled independent studies of the efficacy of LTCI in cats
are required.

Immunoglobulins
In human patients, human intravenous immune globulin (IVIG) FIGURE 7-3  Human intravenous immune globulin solution (inverted for
has been used for treatment of immunodeficiencies, as well as administration).
autoimmune disorders (Figure 7-3). A single infusion of human
IVIG has been used successfully to treat immune-mediated
thrombocytopenia in dogs,74 but repeated administration to Glucocorticoids
animals may lead to transfusion reactions. Intravenous immu- In animals and humans with infectious diseases, treatment with
noglobulin preparations have been commercially available for glucocorticoids is sometimes used to dampen an overzealous
passive immunization of cats or dogs in Europe. These contain immune response that contributes to the pathology of infection.
antibodies to FPV, FHV-1, and FCV (Feliserin, IDT Biologika Glucocorticoids have potent antiinflammatory and immunosup-
GmbH, Rodelben, Germany), or CDV, canine adenovirus, and pressive properties. They suppress cytokine synthesis, decrease
CPV (Stagloban, IDT Biologika GmbH). The efficacy of these antibody production, cause lymphocytolysis and eosinopenia,
preparations has not been reported. Subcutaneous and intraper- decrease neutrophil margination, interfere with cyclooxygen-
itoneal administration of adult cat serum to kittens with failure ase-2, and reduce the activity of phospholipase A2, and thus
of passive transfer can provide IgG concentrations equivalent to initiation of the arachidonic acid cascade. However, glucocor-
those in kittens that suckle normally.75 In general, when given ticoid administration also has the potential to worsen outcome
parenterally in high doses, specific immunoglobulins are only through excessive immune suppression, so their use has been
effective for prevention, and not treatment of an infectious dis- advocated only for specific infectious diseases under certain
ease.76 Oral administration is not effective. Passive administra- circumstances. Glucocorticoids can also contribute to develop-
tion of antitoxins is also used to treat tetanus (see Chapter 54) ment of secondary infections even when they are used to treat
and for neutralization of snake venom components. Passive another disease. For example, it is not unusual in dogs or cats
immunization may be associated with allergic reactions, which for a latent infection to emerge, or for a secondary infection to
can be severe, and it has the potential to interfere with subse- develop (e.g., demodicosis, staphylococcal pyoderma, dermato-
quent active immunization attempts. Administered antibodies phytosis, systemic mycoses) when glucocorticoids are used to
are generally eliminated over a period of 2 to 3 weeks. produce immunosuppression.
An example of the beneficial effect of glucocorticoids for
Lactoferrin treatment of an infectious disease is in FIP, where inflammation
Bovine lactoferrin is a glycoprotein with immunomodulatory plays a major role in pathogenesis. Although controlled clinical
properties that is normally present in exocrine secretions and trials comparing prednisone treatment with a placebo have not
neutrophils. It reduces cytokine secretion by mononuclear cells been performed, for many cats, treatment with prednisolone
and lymphocyte activation in cats infected with FIV. In  vitro, (1 to 2 mg/kg PO q24h) is associated with transient complete
bovine lactoferrin has antiviral activity against FCV and or partial remission and as a result is generally accepted as the
­FHV-1.77,78 Topical application (40 mg/kg q24h for 14 days) to standard of care for treatment of FIP (see Chapter 20).65
the mouth of a small number of FIV-positive and FIV-negative A short course of prednisolone using immunosuppres-
cats with stomatitis was associated with improved appetite and sive doses (1 mg/kg PO q12h) has also been used to suppress
reduced oral inflammation, salivation, and pain, and increased secondary immune-mediated hemolysis in cats infected with
neutrophil phagocytic activity.79 Chronic oral administration of Mycoplasma haemofelis,81 but given that glucocorticoids
bovine lactoferrin to a dog with β2-integrin-related neutrophil can reactivate hemoplasma infections, additional studies are
dysfunction was associated with increased neutrophil function needed to determine whether this practice improves outcome.
and recovery from opportunistic infections.80 Similarly, glucocorticoid treatment has been used to suppress
CHAPTER 7  Antiviral and Immunomodulatory Drugs 63

immune-mediated consequences of infection with the tick-borne In dogs, pentoxifylline has a low and unpredictable oral bio-
pathogens Ehrlichia canis and Babesia spp, when antimicrobial availability and a short half-life, and so three-times-daily
treatment alone is not followed by complete resolution of clini- administration has been recommended. Adverse effects include
cal abnormalities. Adjunctive glucocorticoid treatment did not vomiting and decreased appetite, which are more likely to
worsen outcome in dogs with Rocky Mountain spotted fever.82 occur when the tablets are crushed. Crushed tablets are also
In contrast, glucocorticoid administration may have a negative unpalatable to cats.
impact in dogs with Lyme arthritis.83 There are anecdotal reports
of improved outcome for dogs with suspected Lyme glomerulo- Antimicrobial Drugs with Immunomodulatory Properties
nephritis that are treated with methylprednisolone sodium suc- Antimicrobial drugs with immunomodulatory properties
cinate in conjunction with other immunosuppressive drugs, such include the macrolides, doxycycline, and metronidazole. The
as mycophenolate mofetil or azathioprine (see Chapter 51). mechanisms of these activities are discussed in Chapter 8.
Many large human studies have assessed the role of glucocor-
ticoid treatment in septic shock (see Chapter 86). Treatment with SUGGESTED READINGS
high doses of hydrocortisone for less than 5 days is contraindi- Maggs DJ. Antiviral therapy for feline herpesvirus infections. Vet Clin
cated because of secondary infection and gastrointestinal bleed- North Am Small Anim Pract. 2010;40:1055-1062.
ing. Treatment with physiologic doses of glucocorticoids has been
advocated to overcome relative adrenal insufficiency, which occurs REFERENCES
in severe septic shock in both humans and dogs.84 Recent consen- 1. McDonagh P, Sheehy PA, Norris JM. In  vitro inhibition of
sus guidelines for human patients with septic shock recommend feline coronavirus replication by small interfering RNAs.
adjunctive hydrocortisone treatment in patients who are unre- Vet Microbiol. 2011;150:220-229.
sponsive to fluid resuscitation and treatment with vasopressors.85 2. Nasisse MP, Guy JS, Davidson MG, et al. In vitro suscepti-
bility of feline herpesvirus-1 to vidarabine, idoxuridine, tri-
Infections that generate inflammation within the eye and
fluridine, acyclovir, or bromovinyldeoxyuridine. Am J Vet Res.
CNS are frequently treated with anti-inflammatory doses of 1989;50:158-160.
topical or systemic glucocorticoids, respectively, in conjunc- 3. Williams DL, Fitzmaurice T, Lay L, et al. Efficacy of antiviral
tion with antimicrobial drugs. The goal of such treatment is to agents in feline herpetic keratitis: results of an in vitro study.
minimize or prevent loss of vision or deterioration of neurologic Curr Eye Res. 2004;29:215-218.
status, especially when organism death is followed by a pro- 4. van der Meulen K, Garre B, Croubels S, et al. In vitro com-
found inflammatory response. For CNS infections, the effect of parison of antiviral drugs against feline herpesvirus 1. BMC
concurrent glucocorticoid treatment may depend on the specific Vet Res. 2006;2:13.
pathogen present and the immune response of the infected indi- 5. Malik R, Lessels NS, Webb S, et al. Treatment of feline her-
vidual. Prospective studies that evaluate outcome after adjunc- pesvirus-1 associated disease in cats with famciclovir and
related drugs. J Feline Med Surg. 2009;11:40-48.
tive glucocorticoid treatment in CNS infections of dogs and cats
6. Williams DL, Robinson JC, Lay E, et al. Efficacy of topical
are lacking. In a retrospective study, treatment of dogs and cats acyclovir for the treatment of feline herpetic keratitis: results
with CNS cryptococcosis with antiinflammatory doses of glu- of a prospective clinical trial and data from in vitro investiga-
cocorticoids increased survival in the first 15 days after starting tions. Vet Rec. 2005;157:254-257.
antifungal drug treatment.86 7. Owens JG, Nasisse MP, Tadepalli SM, et al. Pharmacokinetics
of acyclovir in the cat. J Vet Pharmacol Ther. 1996;19:488-490.
Other 8. Nasisse MP, Dorman DC, Jamison KC, et al. Effects of vala-
Pentoxifylline cyclovir in cats infected with feline herpesvirus 1. Am J Vet
Pentoxifylline is a synthetic methylxanthine derived from theo- Res. 1997;58:1141-1144.
bromine. It is a broad-spectrum phosphodiesterase inhibitor 9. Hussein IT, Menashy RV, Field HJ. Penciclovir is a potent
inhibitor of feline herpesvirus-1 with susceptibility deter-
with a variety of immunomodulatory properties. In rodents and
mined at the level of virus-encoded thymidine kinase. Antivi-
humans it inhibits TNF-α, Il-1, Il-6, and Il-12 production, as ral Res. 2008;78:268-274.
well as the activation of T and B cells, and increases neutrophil 10. Thomasy SM, Maggs DJ, Moulin NK, et al. Pharmacokinet-
mobility and chemotaxis. It increases erythrocyte cAMP levels, ics and safety of penciclovir following oral administration of
which results in increased erythrocyte deformability, lowered famciclovir to cats. Am J Vet Res. 2007;68:1252-1258.
blood viscosity, and improved microcirculation and tissue per- 11. Thomasy SM, Lim CC, Reilly CM, et al. Evaluation of orally
fusion. Pentoxifylline also decreases platelet aggregation, and administered famciclovir in cats experimentally infected with
increases tissue plasminogen activator in patients with hyperag- feline herpesvirus type-1. Am J Vet Res. 2011;72:85-95.
gregable platelets, so may be useful for treatment of disorders 12. Thomasy SM, Covert JC, Stanley SD, et al. Pharmacokinetics
that result in hypercoagulable states. The value of pentoxifyl- of famciclovir and penciclovir in tears following oral admin-
istration of famciclovir to cats: a pilot study. Vet Ophthalmol.
line for treatment of endotoxemia has been primarily studied
2012;15:299-306.
in horses. Despite significant decreases in TNF and IL-6 activ- 13. Thomasy SM, Whittem T, Bales J, et  al. Pharmacokinetics of
ity after incubation of equine monocytes with pentoxifylline penciclovir following oral famciclovir and intravenous penciclo-
in vitro, these effects did not occur in horses in vivo.87 Beneficial vir administration in cats. Am J Vet Res. 2012;73(7):1092-1099.
effects of pentoxifylline are limited in horses when administered 14. Maggs DJ, Clarke HE. In vitro efficacy of ganciclovir, cidofo-
IV after challenge with endotoxin.88 vir, penciclovir, foscarnet, idoxuridine, and acyclovir against
In dogs, pentoxifylline has been used to treat a variety of feline herpesvirus type-1. Am J Vet Res. 2004;65:399-403.
skin diseases, including canine atopic dermatitis, dermatomy- 15. Sandmeyer LS, Keller CB, Bienzle D. Effects of cidofovir
ositis, and cutaneous vasculopathies.89,90 In cats it has been on cell death and replication of feline herpesvirus-1 in cul-
used to treat FIP, but a masked, placebo-controlled study of the tured feline corneal epithelial cells. Am J Vet Res. 2005;66:
217-222.
related drug propentofylline showed no benefit of treatment.91
64 SECTION 2  Antiinfective Therapy

16. Fontenelle JP, Powell CC, Veir JK, et al. Effect of topical oph- 36. Centers for Disease Control and Prevention. Guidance on
thalmic application of cidofovir on experimentally induced the use of influenza antiviral agents. 2011. http://www.cdc-
primary ocular feline herpesvirus-1 infection in cats. Am J Vet .gov/flu/professionals/antivirals/antiviral-use-influenza.htm: Last
Res. 2008;69:289-293. accessed May 13, 2012.
17. Stiles J. Treatment of cats with ocular disease attributable to 37. Savigny MR, Macintire DK. Use of oseltamivir in the treat-
herpesvirus infection: 17 cases (1983-1993). J Am Vet Med ment of canine parvoviral enteritis. J Vet Emerg Crit Care
Assoc. 1995;207:599-603. (San Antonio). 2010;20:132-142.
18. Maggs DJ. Antiviral therapy for feline herpesvirus infections. 38. Soong G, Muir A, Gomez MI, et al. Bacterial neuraminidase facil-
Vet Clin North Am Small Anim Pract. 2010;40:1055-1062. itates mucosal infection by participating in biofilm production.
19. Naito T, Irie H, Tsujimoto K, et al. Antiviral effect of arginine J Clin Invest. 2006;116:2297-2305.
against herpes simplex virus type 1. Int J Mol Med. 2009;23: 39. Dal Pozzo F, Galligioni V, Vaccari F, et al. Antiviral efficacy
495-499. of EICAR against canine distemper virus (CDV) in vitro. Res
20. Stiles J, Townsend WM, Rogers QR, et  al. Effect of oral Vet Sci. 2010;88:339-344.
administration of l-lysine on conjunctivitis caused by feline 40. Wyde PR, Moore-Poveda DK, De Clercq E, et al. Use of cot-
herpesvirus in cats. Am J Vet Res. 2002;63:99-103. ton rats to evaluate the efficacy of antivirals in treatment
21. Maggs DJ, Nasisse MP, Kass PH. Efficacy of oral supplemen- of measles virus infections. Antimicrob Agents Chemother.
tation with l-lysine in cats latently infected with feline herpes- 2000;44:1146-1152.
virus. Am J Vet Res. 2003;64:37-42. 41. Yamamoto N, Yang R, Yoshinaka Y, et al. HIV protease inhib-
22. Rees TM, Lubinski JL. Oral supplementation with l-lysine itor nelfinavir inhibits replication of SARS-associated coronavi-
did not prevent upper respiratory infection in a shelter popu- rus. Biochem Biophys Res Commun. 2004;318:719-725.
lation of cats. J Feline Med Surg. 2008;10:510-513. 42. Barnard DL, Day CW, Bailey K, et al. Evaluation of immu-
23. Drazenovich TL, Fascetti AJ, Westermeyer HD, et  al. nomodulators, interferons and known in  vitro SARS-CoV
Effects of dietary lysine supplementation on upper respi- inhibitors for inhibition of SARS-CoV replication in BALB/c
ratory and ocular disease and detection of infectious mice. Antivir Chem Chemother. 2006;17:275-284.
organisms in cats within an animal shelter. Am J Vet Res. 43. Hsieh LE, Lin CN, Su BL, et  al. Synergistic antiviral effect
2009;70:1391-1400. of Galanthus nivalis agglutinin and nelfinavir against feline
24. Maggs DJ, Sykes JE, Clarke HE, et al. Effects of dietary lysine coronavirus. Antiviral Res. 2010;88:25-30.
supplementation in cats with enzootic upper respiratory disease. 44. Tsai HY, Chueh LL, Lin CN, et  al. Clinicopathological
J Feline Med Surg. 2007;9:97-108. findings and disease staging of feline infectious peritonitis:
25. Cattori V, Weibel B, Lutz H. Inhibition of feline leukemia 51 cases from 2003 to 2009 in Taiwan. J Feline Med Surg.
virus replication by the integrase inhibitor raltegravir. Vet 2011;13:74-80.
Microbiol. 2011;152:165-168. 45. Becker AM, Janik TA, Smith EK, et  al. Propionibacterium
26. Greggs 3rd WM, Clouser CL, Patterson SE, et al. Discovery acnes immunotherapy in chronic recurrent canine pyo-
of drugs that possess activity against feline leukemia virus. J derma. An adjunct to antibiotic therapy. J Vet Intern Med.
Gen Virol. 2012;93:900-905. 1989;3:26-30.
27. Gomez NV, Castillo VA, Gisbert MA, et al. Immune-endocrine 46. Yates KM, Rosenberg LJ, Harris CK, et al. Pilot study of the
interactions in treated and untreated cats naturally infected effect of acemannan in cats infected with feline immunodefi-
with FIV. Vet Immunol Immunopathol. 2011;143:332-337. ciency virus. Vet Immunol Immunopathol. 1992;35:177-189.
28. Hartmann K, Donath A, Beer B, et al. Use of two virustatica 47. Sheets MA, Unger BA, Giggleman Jr GF, et al. Studies of the
(AZT, PMEA) in the treatment of FIV and of FeLV sero- effect of acemannan on retrovirus infections: clinical stabi-
positive cats with clinical symptoms. Vet Immunol Immuno- lization of feline leukemia virus-infected cats. Mol Biother.
pathol. 1992;35:167-175. 1991;3:41-45.
29. Nelson P, Sellon R, Novotney C, et al. Therapeutic effects of 47a. Legendre A. Personal communication. 2011.
diethylcarbamazine and 3’-azido-3’-deoxythymidine on feline 48. Legendre AM, Bartges JW. Effect of polyprenyl immunostimu-
leukemia virus lymphoma formation. Vet Immunol Immuno- lant on the survival times of three cats with the dry form of feline
pathol. 1995;46:181-194. infectious peritonitis. J Feline Med Surg. 2009;11:624-626.
30. Fogle JE, Tompkins WA, Campbell B, et al. Fozivudine tidoxil 49. Page AV, Liles WC. Immunomodulators. In: Mandell GL,
as single-agent therapy decreases plasma and cell-associated Bennett JE, Dolin R, eds. Principles and Practice of Infectious
viremia during acute feline immunodeficiency virus infection. Diseases. 7th ed. Philadelphia, PA: Churchill Livingstone,
J Vet Intern Med. 2011;25:413-418. Elsevier; 2010:611-623.
31. Arai M, Earl DD, Yamamoto JK. Is AZT/3TC therapy 50. Schefold JC. Immunostimulation using granulocyte- and
effective against FIV infection or immunopathogenesis? Vet granulocyte-macrophage colony stimulating factor in patients
Immunol Immunopathol. 2002;85:189-204. with severe sepsis and septic shock. Crit Care. 2011;15:136.
32. Martinez-Ruzafa I, Dominguez PA, Dervisis NG, et al. Toler- 51. Cheng AC, Stephens DP, Currie BJ. Granulocyte-colony stimu-
ability of gemcitabine and carboplatin doublet therapy in cats lating factor (G-CSF) as an adjunct to antibiotics in the treatment
with carcinomas. J Vet Intern Med. 2009;23:570-577. of pneumonia in adults. Cochrane Database Syst Rev. 2007:
33. Hartmann K, Stengel C, Klein D, et al. Efficacy and adverse CD004400.
effects of the antiviral compound plerixafor in feline immu- 52. Fulton R, Gasper PW, Ogilvie GK, et al. Effect of recombi-
nodeficiency virus-infected cats. J Vet Intern Med. 2012;26: nant human granulocyte colony-stimulating factor on hema-
483-490. topoiesis in normal cats. Exp Hematol. 1991;19:759-767.
34. Lascelles BD, Gaynor JS, Smith ES, et  al. Amantadine in 53. Rewerts JM, McCaw DL, Cohn LA, et  al. Recombinant
a multimodal analgesic regimen for alleviation of refrac- human granulocyte colony-stimulating factor for treatment
tory osteoarthritis pain in dogs. J Vet Intern Med. 2008;22: of puppies with neutropenia secondary to canine parvovirus
53-59. infection. J Am Vet Med Assoc. 1998;213:991-992.
35. Li W, Escarpe PA, Eisenberg EJ, et  al. Identification of GS 54. Mischke R, Barth T, Wohlsein P, et  al. Effect of recombi-
4104 as an orally bioavailable prodrug of the influenza virus nant human granulocyte colony-stimulating factor (rhG-CSF)
neuraminidase inhibitor GS 4071. Antimicrob Agents Che- on leukocyte count and survival rate of dogs with parvoviral
mother. 1998;42:647-653. enteritis. Res Vet Sci. 2001;70:221-225.
CHAPTER 7  Antiviral and Immunomodulatory Drugs 65

55. Phillips K, Arai M, Tanabe T, et al. FIV-infected cats respond 73. Hampel V, Schwarz B, Kempf C, et al. Adjuvant immunother-
to short-term rHuG-CSF treatment which results in anti-G- apy of feline fibrosarcoma with recombinant feline interferon-
CSF neutralizing antibody production that inactivates drug omega. J Vet Intern Med. 2007;21:1340-1346.
activity. Vet Immunol Immunopathol. 2005;108:357-371. 74. Bianco D, Armstrong PJ, Washabau RJ. A prospective, ran-
56. Yamamoto A, Iwata A, Saito T, et al. Expression and puri- domized, double-blinded, placebo-controlled study of human
fication of canine granulocyte colony-stimulating factor (cG- intravenous immunoglobulin for the acute management of
CSF). Vet Immunol Immunopathol. 2009;130:221-225. presumptive primary immune-mediated thrombocytopenia in
57. Yamamoto A, Fujino M, Tsuchiya T, et  al. Recombinant dogs. J Vet Intern Med. 2009;23:1071-1078.
canine granulocyte colony-stimulating factor accelerates 75. Levy JK, Crawford PC, Collante WR, et al. Use of adult cat
recovery from cyclophosphamide-induced neutropenia in serum to correct failure of passive transfer in kittens. J Am
dogs. Vet Immunol Immunopathol. 2011;142:271-275. Vet Med Assoc. 2001;219:1401-1405.
58. Duffy A, Dow S, Ogilvie G, et al. Hematologic improvement 76. Day MJ, Horzinek MC, Schultz RD. WSAVA guidelines
in dogs with parvovirus infection treated with recombinant for the vaccination of dogs and cats. J Small Anim Pract.
canine granulocyte-colony stimulating factor. J Vet Pharma- 2010;51:1-32.
col Ther. 2010;33:352-356. 77. McCann KB, Lee A, Wan J, et al. The effect of bovine lacto-
59. Arai M, Darman J, Lewis A, et al. The use of human hema- ferrin and lactoferricin B on the ability of feline calicivirus (a
topoietic growth factors (rhGM-CSF and rhEPO) as a norovirus surrogate) and poliovirus to infect cell cultures. J
supportive therapy for FIV-infected cats. Vet Immunol Immu- Appl Microbiol. 2003;95:1026-1033.
nopathol. 2000;77:71-92. 78. Beaumont SL, Maggs DJ, Clarke HE. Effects of bovine lacto-
60. Mayer P, Werner FJ, Lam C, et al. In vitro and in vivo activity ferrin on in vitro replication of feline herpesvirus. Vet Oph-
of human recombinant granulocyte-macrophage colony-stim- thalmol. 2003;6:245-250.
ulating factor in dogs. Exp Hematol. 1990;18:1026-1033. 79. Sato R, Inanami O, Tanaka Y, et al. Oral administration of
61. Weiss RC, Cox NR, Oostrom-Ram T. Effect of interferon bovine lactoferrin for treatment of intractable stomatitis in
or Propionibacterium acnes on the course of experimentally feline immunodeficiency virus (FIV)-positive and FIV-nega-
induced feline infectious peritonitis in specific-pathogen-free tive cats. Am J Vet Res. 1996;57:1443-1446.
and random-source cats. Am J Vet Res. 1990;51:726-733. 80. Kobayashi S, Abe Y, Inanami O, et al. Oral administration of
62. Zeidner NS, Myles MH, Mathiason-DuBard CK, et al. Alpha bovine lactoferrin upregulates neutrophil functions in a dog
interferon (2b) in combination with zidovudine for the treat- with familial beta2-integrin-related neutrophil dysfunction.
ment of presymptomatic feline leukemia virus-induced immu- Vet Immunol Immunopathol. 2011;143:155-161.
nodeficiency syndrome. Antimicrob Agents Chemother. 81. Tasker S, Lappin MR. Haemobartonella felis: recent devel-
1990;34:1749-1756. opments in diagnosis and treatment. J Feline Med Surg.
63. Penzo C, Ross M, Muirhead R, et al. Effect of recombinant 2002;4:3-11.
feline interferon-omega alone and in combination with che- 82. Breitschwerdt EB, Davidson MG, Hegarty BC, et al. Predniso-
motherapeutic agents on putative tumour-initiating cells lone at anti-inflammatory or immunosuppressive dosages in
and daughter cells derived from canine and feline mammary conjunction with doxycycline does not potentiate the severity
tumours. Vet Comp Oncol. 2009;7:222-229. of Rickettsia rickettsii infection in dogs. Antimicrob Agents
64. Ishida T, Shibanai A, Tanaka S, et  al. Use of recombinant Chemother. 1997;41:141-147.
feline interferon and glucocorticoid in the treatment of feline 83. Chang YF, Novosel V, Chang CF, et al. Experimental induc-
infectious peritonitis. J Feline Med Surg. 2004;6:107-109. tion of chronic borreliosis in adult dogs exposed to Borrelia
65. Ritz S, Egberink H, Hartmann K. Effect of feline interferon- burgdorferi-infected ticks and treated with dexamethasone.
omega on the survival time and quality of life of cats with feline Am J Vet Res. 2001;62:1104-1112.
infectious peritonitis. J Vet Intern Med. 2007;21:1193-1197. 84. Peyton JL, Burkitt JM. Critical illness-related corticosteroid
66. de Mari K, Maynard L, Sanquer A, et al. Therapeutic effects insufficiency in a dog with septic shock. J Vet Emerg Crit Care
of recombinant feline interferon-omega on feline leukemia (San Antonio). 2009;19:262-268.
virus (FeLV)-infected and FeLV/feline immunodeficiency 85. Dellinger RP, Levy MM, Carlet JM, et al. Surviving Sepsis Cam-
virus (FIV)-coinfected symptomatic cats. J Vet Intern Med. paign: international guidelines for management of severe sepsis
2004;18:477-482. and septic shock: 2008. Crit Care Med. 2008;36:296-327.
67. Domenech A, Miro G, Collado VM, et al. Use of recombinant 86. Sykes JE, Sturges BK, Cannon MS, et al. Clinical signs, imag-
interferon omega in feline retrovirosis: from theory to prac- ing features, neuropathology, and outcome in cats and dogs
tice. Vet Immunol Immunopathol. 2011;143:301-306. with central nervous system cryptococcosis from California. J
68. Hennet PR, Camy GA, McGahie DM, et  al. Comparative Vet Intern Med. 2010;24:1427-1438.
efficacy of a recombinant feline interferon omega in refrac- 87. Baskett A, Barton MH, Norton N, et  al. Effect of pent-
tory cases of calicivirus-positive cats with caudal stomatitis: oxifylline, flunixin meglumine, and their combination on a
a randomised, multi-centre, controlled, double-blind study in model of endotoxemia in horses. Am J Vet Res. 1997;58:
39 cats. J Feline Med Surg. 2011;13:577-587. 1291-1299.
69. Haid C, Kaps S, Gonczi E, et  al. Pretreatment with feline 88. Barton MH, Moore JN, Norton N. Effects of pentoxifylline
interferon omega and the course of subsequent infection with infusion on response of horses to in vivo challenge exposure
feline herpesvirus in cats. Vet Ophthalmol. 2007;10:278-284. with endotoxin. Am J Vet Res. 1997;58:1300-1307.
70. Southerden P, Gorrel C. Treatment of a case of refractory 89. Marsella R, Olivry T. The ACVD task force on canine atopic
feline chronic gingivostomatitis with feline recombinant inter- dermatitis (XXII): nonsteroidal anti-inflammatory pharmaco-
feron omega. J Small Anim Pract. 2007;48:104-106. therapy. Vet Immunol Immunopathol. 2001;81:331-345.
71. Gutzwiller ME, Brachelente C, Taglinger K, et al. Feline her- 90. Nichols PR, Morris DO, Beale KM. A retrospective study
pes dermatitis treated with interferon omega. Vet Dermatol. of canine and feline cutaneous vasculitis. Vet Dermatol.
2007;18:50-54. 2001;12:255-264.
72. Paltrinieri S, Crippa A, Comerio T, et al. Evaluation of inflam- 91. Fischer Y, Ritz S, Weber K, et al. Randomized, placebo con-
mation and immunity in cats with spontaneous parvovirus trolled study of the effect of propentofylline on survival time
infection: consequences of recombinant feline interferon-omega and quality of life of cats with feline infectious peritonitis. J
administration. Vet Immunol Immunopathol. 2007;118:68-74. Vet Intern Med. 2011;25:1270-1276.
CHAPTER 8

Antibacterial Drugs
Jane E. Sykes and Mark G. Papich

w KEY POINTS
• A
 n understanding of the mechanisms of action, spectrum of • T he importance of appropriate administration of antibiotics,
activity, pharmacokinetics, pharmacodynamics, and adverse compliance issues, and potential adverse effects of the drug
effects of antibacterial drugs facilitates optimal treatment of bac- chosen should be discussed with the pet owner when antibiot-
terial infections while minimizing the chance for (1) selection of ics are prescribed.
resistant bacteria, (2) adverse reactions, and (3) drug interactions.

INTRODUCTION action of β-lactam antibiotics. For example, inhibition of PBP1a


and PBP1b leads to cell lysis, whereas inhibition of PBP2 results
This chapter reviews the classification, mechanisms of action, in rounded cells called spheroblasts. Drugs that produce rapid
spectrum of activity, resistance mechanisms, tissue penetration, lysis (e.g., carbapenems) are the most bactericidal and have
clinical use, and adverse effects of the major classes of antibiot- highest affinity for PBP1.
ics used to treat dogs and cats. Antimicrobials have been grouped Resistance to β-lactam antibiotics is now widespread and
together alphabetically according to their mechanism of action results primarily from β-lactamase production.2 It can also
(cell wall synthesis, nucleic acid, and then protein synthesis inhibi- result from production of altered PBPs (such as PBP2a) and,
tors). Tables are dispersed throughout the text with drug dosages among gram-negative bacteria, exclusion of drugs that normally
for each group of antibiotics. The reader is encouraged to review diffuse through porins to their site of action. Gram-negative
the accompanying text when using the tables. Not all antibiotics β-lactamases are strategically located just beneath the outer
are reviewed here. Antibiotics that have specific uses, such as cer- lipopolysaccharide layer, which acts as the barrier to drug pen-
tain antimycobacterial drugs and nitrofurantoin, are reviewed in etration. Gram-positive bacteria secrete β-lactamases into their
the relevant chapters of this book. Chapter 6 provides an overview immediate surroundings. There are many different β-lactamase
of the basic principles of antimicrobial treatment. enzymes that vary in their specificity for β-lactam drugs.3
β-Lactam antibiotics have a short half-life and exhibit time-
Inhibitors of Cell Wall Synthesis dependent pharmacodynamics (see Chapter 6). Drug concen-
trations should be maintained above the minimum inhibitory
Beta-Lactam Antibiotics concentration (T > MIC) for at least 50% of the dosing interval.
β-Lactam antibiotics have a β-lactam ring in their molecular For penicillins and carbapenems, T > MIC can be less than 50%,
structure (Figure 8-1). They include penicillins, cephalosporins, but aiming for a target of 50% ensures that most patients will
monobactams, and carbapenems. They are bactericidal antibiot- have adequate exposure. To maintain this target, some β-lactam
ics (see Chapter 6) that bind covalently to and inhibit penicillin antibiotics with short half-lives require frequent administration
binding proteins (PBPs). These PBPs are needed to catalyze the or slow infusion. For other drugs, a long half-life prolongs the
cross-linking (or transpeptidation) of the peptidoglycan layer of
bacterial cell walls, which is continuously remodeled by bacteria
(Figure 8-2). When PBPs are inactivated by β-lactam antibiotics,
bacterial enzymes that hydrolyze the peptidoglycan cross-links
during cell wall remodeling continue to function, which breaks
down the cell wall further. The accumulation of peptidoglycan
precursors also triggers activation of cell wall hydrolases, with
further digestion of intact peptidoglycan. The end result is bac-
terial rupture.
The peptidoglycan layer of gram-positive bacteria is 50 to
100 times thicker than that of gram-negative bacteria and is
highly cross-linked, which maintains the structural integrity of
gram-positive bacteria.1 Therefore, compared to gram-negative
bacteria, gram-positive bacteria are more easily inactivated by
β-lactam antibiotics. Bacteria possess multiple PBPs, which are
assigned numbers based on their molecular weight. PBPs vary in
their affinities for different β-lactam antibiotics, which in part
explains the difference in spectrum of activity and bactericidal FIGURE 8-1  Structure of β-lactam antibiotics.

66
CHAPTER 8  Antibacterial Drugs 67

T > MIC to allow for infrequent administration. For example, 14-day intervals. For other cephalosporins (e.g., injectable drugs
cefpodoxime proxetil has a half-life longer than those of other with short half-lives such as cefotaxime), three- to four-times-
oral cephalosporins, so it can be administered only once per daily dosing may be required for treatment of gram-negative
day. Cefovecin has an extremely long half-life in dogs and cats bacterial infections.
and can be effective for some infections when administered at
Penicillins
Classification, Spectrum of Activity, and Resistance Mechanisms
Penicillins are derived from Penicillium molds. They are clas-
sified as naturally occurring penicillins such as penicillin G
(benzyl penicillin), the semisynthetic aminopenicillins such as
ampicillin and amoxicillin, penicillinase-resistant penicillins,
A and extended-spectrum penicillins. Table 8-1 shows the classifi-
cation of the penicillins and their respective spectra of activity.
Penicillin G must be given parenterally because of poor
oral bioavailability. Gram-positive bacteria that are suscep-
B tible to penicillin G are more susceptible to penicillin G than
aminopenicillins such as ampicillin. The aminopenicillins have
some activity against gram-negative bacteria, although many
gram-negative bacteria are resistant because they produce
β-lactamases. Ampicillin has limited absorption from the gas-
C
trointestinal tract, so it should be given parenterally for treat-
ment of systemic infections. Amoxicillin has approximately
twice the oral absorption of ampicillin and can be given orally.
Penicillinase-resistant penicillins (antistaphylococcal penicil-
D lins) such as methicillin have a structure that resists inactivation
by β-lactamase enzymes. Methicillin is no longer used clinically.
Drugs such as flucloxacillin and dicloxacillin have replaced the
use of methicillin, because they can be administered orally and
E have fewer adverse effects. These drugs are highly protein-bound,
which delays their elimination and maintains plasma drug con-
FIGURE 8-2  Interference of peptidoglycan production by β-lactam antibiotics. The
bacterial cell wall consists of N-acetylglucosamine (NAG) and N-acetylmuramic acid (NAM) centrations, but tissue penetration is sufficient for treatment of
subunits. A, The penicillin binding protein (PBP, brown squares) binds peptide side chains skin and soft-tissue infections. Although dosages of penicillinase-
and forms a cross-link (B). C, The PBP dissociates from the wall once the cross-link has been resistant penicillins have been published for dogs and cats,4 they
formed. D, Penicillins enter the active site of the PBP. E, The β-lactam ring of the penicillin is are rarely used because other alternatives exist such as the com-
irreversibly opened when it reacts with the PBP, which permanently blocks the active site. bination of amoxicillin and the β-lactamase inhibitor clavulanic

TABLE 8-1
Classification and Spectrum of Activity of Penicillin Antibiotics
Penicillin Group Examples Antibacterial Spectrum
Natural penicillins Penicillin G Gram-positive aerobic bacteria (especially streptococci) and anaer-
obes. Pasteurella multocida may also be susceptible.
Aminopenicillins Ampicillin Gram-positive aerobic bacteria (cocci and bacilli) and anaerobes;
Amoxicillin some susceptible gram-negative bacteria.
Penicillinase-resistant Methicillin Susceptible β-lactamase producing gram-positive aerobic bacte-
­penicillins Oxacillin ria. Less active against penicillinase-susceptible organisms than
Cloxacillin penicillin G.
Dicloxacillin
Extended spectrum Ticarcillin Increased activity against susceptible gram-negative aerobes, which
­carboxypenicillins Carbenicillin includes Pseudomonas aeruginosa, Proteus spp., Klebsiella, and
anaerobes. Some reduction in gram-positive spectrum. De-
stroyed by β-lactamases.
Ureidopenicillins Mezlocillin Susceptible gram-positive and gram-negative aerobes, which
Piperacillin includes Pseudomonas aeruginosa. Some loss of anaerobic spec-
trum. Destroyed by β-lactamases.
Penicillin–β-lactamase β-Lactamase inhibitors: Susceptible β-lactamase producing gram-positive and gram negative
inhibitor combinations clavulanic acid, sulbactam, aerobic bacteria and anaerobes.
tazobactam
68 SECTION 2  Antiinfective Therapy

acid. Methicillin and oxacillin are used in the laboratory for sus- Because of low penetration across the blood-brain barrier and
ceptibility testing. Resistance to methicillin results from bacte- blood-ocular barrier, high doses are needed in order to attain
rial production of an altered penicillin-binding protein (PBP2a), effective levels in these tissues. Penicillins undergo rapid renal
which leads to resistance to all β-lactam antibiotics.5 elimination, and active drug is concentrated in the urine, so
Extended-spectrum penicillins (antipseudomonal penicil- they are useful for treatment of bacterial urinary tract infections
lins) have enhanced activity against gram-negative bacteria, (UTIs). Liver impairment has little effect on the pharmacokinet-
but must be given parenterally. They include the carboxypeni- ics of penicillins. Increasing the length of the dose interval is
cillins, such as ticarcillin and carbenicillin, and piperacillin, an important in order to minimize toxicity in animals with renal
ureidopenicillin. failure.
Clavulanic acid, sulbactam, and tazobactam are β-lactamase Because of a short half-life, frequent dosing is necessary,
inhibitors that are administered in conjunction with penicillins, especially when treating gram-negative bacterial infections with
which increases the spectrum of activity. β-Lactamase inhibitors aminopenicillins. Penicillin MICs are generally higher for gram-
possess weak intrinsic antibacterial activity. Clavulanate has negative bacteria than gram-positive bacteria; therefore, higher
been combined not only with amoxicillin (oral), but also ticar- penicillin dosages may also be needed for gram-negative bacte-
cillin (parenteral). Sulbactam has been combined with ampicil- rial infections.
lin for intravenous (IV) injection. Sulbactam is not as active as
clavulanic acid against some gram-negative β-lactamases. Tazo- Adverse Effects
bactam is combined with piperacillin. Penicillins are generally well tolerated by dogs and cats. The
most common adverse effects in dogs and cats are vomiting,
Clinical Use diarrhea, and inappetence. These are more likely to occur
Penicillin dosages for dogs and cats are listed in Table 8-2. Peni- with amoxicillin when clavulanic acid is administered concur-
cillins are distributed to the extracellular fluid of most tissues. rently. Hypersensitivity reactions that include fever, cutaneous

TABLE 8-2
Suggested Penicillin Dosages for Small Animals
Dose Interval CLSI Breakpoints
Drug (mg/kg) (hours) Species Route Comments (µg/mL)
Penicillin G ­ 20,000-40,000 6-8 D, C IV, IM ≤8 enterococci;
potassium or U/kg ≤0.12 staphylococci
­sodium and streptococci
Ampicillin sodium 10-20 6-8 D, C IV, IM, Administer IV over 3 minutes. ≤0.25 staphylococci,
SC When reconstituted with streptococci, and
sterile water to 250 mg/mL, gram-negative bacilli;
vials should be used within ≤8 canine urinary tract
1 hour. When reconstituted pathogens
with 0.9% NaCl to 45 mg/
mL, stability is maintained
for 48 hours if refrigerated.
Ampicillin-­ 10-20 8 D, C IV, IM See ampicillin. Dose according ≤8/4* staphylococci and
sulbactam to ampicillin component. gram-negative bacteria
Amoxicillin 10-20 8-12 D, C PO See ampicillin.
Amoxicillin-­ 12.5-25 12 D, C PO More frequent or higher ≤0.25/0.12* staphylo-
clavulanate doses recommended for cocci, streptococci,
­potassium ­gram-negative infections. Escherichia coli and
Pasteurella multocida
Ticarcillin ­disodium, 33-50 4-6 D, C IV When reconstituted with ≤64/2* Pseudomonas
­ticarcillin- 0.9% NaCl, stable for up to aeruginosa
clavulanate† 14 days when refrigerated. ≤16/2 gram-negative
enteric bacteria
Piperacillin ­sodium/ 40 6 D, C IV Reconstituted solution should See ticarcillin.
piperacillin-­ be used within 7 days if
tazobactam† ­refrigerated.

C, Cats; CLSI, Clinical and Laboratory Standards Institute; D, dogs.


*The “/” distinguishes the ticarcillin from clavulanic acid concentrations.
†It is recommended that the use of these drugs be reserved for life-threatening infections that are susceptible to these drugs but resistant to other drugs,

on the basis of culture and susceptibility testing.


CHAPTER 8  Antibacterial Drugs 69

reactions, immune-mediated cytopenias, and anaphylaxis are urine, and so are useful for treatment of UTIs. Cefazolin has
possible, especially in dogs, but are less common than in human greater activity against a wider range of bacterial isolates than
patients. The use of penicillins should be avoided in animals the oral first-generation cephalosporins (e.g., cephalexin and
with a history of such reactions. Rapid intravenous infusion cefadroxil).7 Cefazolin has been a popular injectable antibiotic
or administration of high doses of penicillins to animals with for perioperative use in small animals. This perioperative use of
coexisting renal failure is rarely followed by development of cefazolin is with the intention to prevent postsurgical infections,
neurologic signs, such as seizures. Pain or tissue reactions can particularly in orthopedic surgery (see Chapter 85).
occur when penicillins are administered intramuscularly or sub- Use of second-, third-, and fourth-generation cephalosporins
cutaneously. High doses of extended-spectrum penicillins such may be indicated when there is resistance to first-generation
as ticarcillin have been associated with platelet function abnor- cephalosporins and there are no other reasonable alternatives
malities and significant bleeding in human patients.6 available. Three third-generation cephalosporins are approved
by the U.S. Food and Drug Administration (FDA) for use in
Cephalosporins small animals: ceftiofur, cefpodoxime proxetil, and cefovecin.
Classification and Spectrum of Activity These agents are approved for treatment of skin and soft-tissue
The cephalosporins were originally derived from Acremonium infections (cefpodoxime proxetil and cefovecin) and UTIs in
spp. (previously known as Cephalosporium spp.). They have dogs (ceftiofur). Although their use in small animal medicine
been broadly grouped into first-, second-, third-, and fourth- has been controversial because of concerns that relate to selec-
generation cephalosporins based on their spectrum of activity. tion for resistant bacteria, there is no evidence at this time that
As the generation increases, there is an increase in gram-negative the use of these agents has been associated with increased resis-
spectrum, and fourth-generation drugs have truly broad-spectrum tance among isolates from dogs and cats. The injectable third-
activity (Table 8-3). An exception to this pattern is the new- generation cephalosporins approved for people have greater in
est addition to the group, ceftaroline. It has activity against vitro activity than cefpodoxime or cefovecin and occasionally
­methicillin-resistant staphylococci as a result of an affinity for have been used in dogs and cats (e.g., cefotaxime, ceftazidime,
PBP2a and has been considered a “fifth-generation” cephalospo- cefoperazone). These drugs have a high degree of activity, but
rin. Cephalosporins are resistant to staphylococcal β-lactamase, are expensive and must be administered frequently by injec-
and the extended-spectrum cephalosporins (primarily the tion. Distribution and excretion into urine and bile is variable.
third-generation drugs) are resistant to many gram-negative Some third-generation cephalosporins have a longer duration
β-lactamases. However, extended-spectrum β-lactamase (ESBL) of activity than first- and second-generation drugs by virtue of
enzymes can hydrolyze even third-generation cephalosporins long half-lives. Cefpodoxime proxetil is the most commonly
and present an important therapeutic challenge. used oral third-generation drug in small animals, approximately
50% of which is excreted in the urine in active form. Cefove-
Clinical Use cin is approved for use in dogs and cats in Europe and North
Cephalosporin dosages for cats and dogs are listed in Table 8-4. America as a subcutaneous injection. The high protein bind-
First-generation cephalosporins such as cephalexin are moder- ing and slow clearance result in a very long half-life in these
ately absorbed orally and distributed into extracellular fluids. In species (approximately 7 days in cats and 5 days in dogs).
general, they have poor penetration of blood-brain barrier, even Administration of cefovecin to cats results in a duration of
in the face of inflammation. Most are excreted unchanged in the activity of at least 14 days, and for at least 7 to 14 days in dogs

TABLE 8-3
Classification and Spectrum of Activity of Cephalosporin Antibiotics
Cephalosporin
­Generation Examples Antibacterial Spectrum
First Cephalexin Methicillin-susceptible staphylococci and streptococci and aerobes; some activity against
Cefazolin gram-negative aerobes (especially cefazolin). Unpredictable activity against anaerobes.
Cefadroxil
Second Cefaclor Improved activity against gram-negative aerobes and anaerobes compared with first-
Cefotetan generation drugs. Cefoxitin has greater stability against anaerobic β-lactamases (such
Cefoxitin as those produced by Bacteroides) than other second-generation drugs.
Third Cefixime Activity primarily against gram-negative bacteria. Some drugs in this group have less
Cefpodoxime activity against gram-positive bacteria and anaerobes.
Ceftiofur
Cefotaxime
Ceftriaxone
Cefovecin
Ceftazidime
Fourth Cefepime Gram-positive cocci, gram-negative bacilli, many β-lactamase-producing gram-negative
bacteria. Reserve for life-threatening infections where no other alternative is available.
70 SECTION 2  Antiinfective Therapy

TABLE 8-4
Suggested Cephalosporin Dosages for Small Animals
Dose Interval
Drug (mg/kg) (hours) Route Species Comments CLSI Breakpoints (µg/mL)
Cephalexin 10-30 6-12 PO D Use 30 mg/kg q12h for pyoderma. ≤2 susceptible organisms.
15-20 12 C More frequent dosing (q6-8h) Cephalothin can be used
is required for gram-negative to test susceptibility for all
bacterial infections. other 1st generation drugs.
Cefadroxil 22 12 PO D See cephalexin.
24 C
Cefazolin sodium 20-35 8 IV, IM D, C 22 mg/kg q2h during surgery See cephalexin.
if indicated.
Cefaclor† 15-20 8 PO D, C ≤8 all susceptible organisms
Cefoxitin sodium† 30 6-8 IM, IV D, C Pain on injection IM. See cefaclor.
Cefixime† 10 12 PO D, C Use 5 mg/kg q12-24h for ≤1 all susceptible organisms
urinary tract infections.
Cefpodoxime proxetil† 5-10 24 PO D* Of all third-generation cephalo- ≤2 all susceptible organisms
sporins, most active against
staphylococci. Less active
against gram-negative and
anaerobic infections.
Ceftiofur sodium† 2.2-4.4 24 SC D* For treatment of otherwise resistant Not established for dogs
urinary tract ­infections. and cats
Cefotaxime sodium† 50 12 IV, IM D Partially metabolized by the liver See cefpodoxime.
20-80 6 C to desacetyl cefotaxime which
contributes to antibacterial
activity. Good CNS penetra-
tion. Stable for 5 days in the
refrigerator after reconstitution.
Ceftazidime† 30 6 IV D, C Potent antipseudomonal ­activity. ≤4 Enterobacteriaceae, ≤8
Pseudomonas aeruginosa
and staphylococci
Cefovecin† 8 14 days SC D, C Approved for use for skin and soft No approved breakpoint.
tissue infections and in some ≤2 all susceptible organ-
countries, urinary tract infec- isms is suggested
tions. Efficacy for treatment
of infections in other sites not
established.
Cefepime† 40 6 IV, IM D* Reduce frequency in dogs with ≤ 8 all susceptible organisms
renal failure (q12-24h).

C, Cats; CLSI, Clinical and Laboratory Standards Institute; CNS, central nervous system; D, dogs.
*Dose not established for cats.
†It is recommended that the use of most second-, third-, and fourth-generation cephalosporins be reserved for serious infections that are susceptible

to those drugs but resistant to other reasonable alternatives on the basis of culture and susceptibility testing.

(depending on the pathogen). Injections can be repeated in dogs Cephalosporins have the potential to cause false-positive results
at 14-day intervals if necessary. on test strips that use copper reduction for urine glucose detec-
tion. Certain cephalosporins, such as cefotetan and ceftriaxone,
Adverse Effects may exacerbate bleeding tendencies due to vitamin K antago-
Adverse effects of cephalosporins in dogs and cats are similar to nism.8 Reversible bone marrow suppression has been reported
those of penicillins, with gastrointestinal signs and hypersensitiv- in dogs given high doses of ceftiofur, cefonicid, and cefazedone
ity reactions being most frequent. Although the incidence is low, long-term.9
cross-sensitivity can occur between penicillins and cephalospo-
rins, so cephalosporins should be avoided or used with caution Monobactams (Aztreonam)
in animals that have a history of reactions to penicillin. Inject- Aztreonam is a synthetic β-lactam antibiotic that is resistant
able cephalosporins can cause phlebitis and pain on injection. to some β-lactamases. It is effective against a wide range of
CHAPTER 8  Antibacterial Drugs 71

TABLE 8-5
Suggested Carbapenem Dosages for Small Animals
Dose Interval CLSI Breakpoints
Drug (mg/kg) (hours) Species Route Comments (µg/mL)
Imipenem-­ 5 6-8 D, C IV, IM, SC 250 to 500 mg of reconstituted drug ≤1 Enterobac-
cilastatin should be added to no less than teriaceae, ≤2
100 mL of fluids and administered Pseudomonas
IV over 30-60 minutes. Stable for aeruginosa
24 hours after reconstitution when
refrigerated. For IM injections,
reconstitute with 1% lidocaine.
Meropenem 8.5 12 D, C SC SC injections may cause alopecia See imipenem.
24 12 D, C IV at the injection site. For Pseudo-
monas aeruginosa or infections
with MIC values approaching the
breakpoint, use 12 mg/kg q8h SC
or 24 mg/kg q8h IV.

Carbapenems should be used for serious infections resistant to other reasonable alternatives, on the basis of culture and susceptibility testing.
C, Cats; CLSI, Clinical and Laboratory Standards Institute; D, dogs; MIC, minimum inhibitory concentration.

gram-negative bacteria, including Pseudomonas aeruginosa, but Adverse Effects


not gram-positive or anaerobic bacteria. In human medicine, Imipenem is degraded by dehydropeptidase-1, a brush border
aztreonam is used to treat patients with serious gram-negative enzyme in the proximal renal tubules, which results in produc-
infections who are allergic to penicillins and cannot tolerate tion of an inactive metabolite that is nephrotoxic. In order to
aminoglycosides.10 Dosages have not been established for dogs. prevent nephrotoxicity and maximize imipenem’s antibacterial
activity, imipenem is administered with cilastatin, which inhib-
Carbapenems its the dehydropeptidase-1 enzyme. Newer carbapenems such as
Spectrum of Activity and Clinical Use meropenem are resistant to dehydropeptidase-1.
Carbapenems are highly resistant to almost all bacterial Adverse effects of carbapenems are otherwise rare and
β-lactamases and include imipenem, meropenem, ertapenem, include vomiting, nausea, and pain on injection. Neurologic
and doripenem. They penetrate the outer membrane of gram- signs, including tremors, nystagmus, and seizures, can occur
negative bacteria more effectively than many other β-lactam following rapid infusion of imipenem-cilastatin or in animals
antibiotics and bind to a variety of PBPs, which leads to rapid with renal insufficiency. Imipenem must be administered slowly
lysis of a broad spectrum of bacteria.11 In contrast to other in intravenous fluids (see Table 8-5). Slow administration is not
β-lactam antibiotics, carbapenems have a postantibiotic effect required for meropenem, which is more soluble. Subcutaneous
(see Chapter 6) similar to that of aminoglycosides and fluo- injections of meropenem have produced alopecia at the injec-
roquinolones. Although effective against many gram-positive, tion site,4 but otherwise these injections have been very well
gram-negative, and anaerobic bacterial infections, the use tolerated and have eradicated infections associated with MDR
of carbapenems is reserved for treatment of serious, multiple gram-negative bacteria.
drug-resistant (MDR) gram-negative infections, especially Esch-
erichia coli, Klebsiella pneumoniae, and P. aeruginosa, that are Glycopeptides
resistant to other antibiotics. Methicillin-resistant staphylococci Mechanism of Action and Clinical Use
are resistant to carbapenems. Dosages for dogs and cats are Glycopeptides are cyclic glycosylated peptide antimicrobials
listed in Table 8-5. that inhibit the synthesis of peptidoglycan by binding to amino
The identification of carbapenemase production by some acids (d-alanyl-d-alanine) in the cell wall, preventing the addi-
strains of gram-negative bacteria has raised great concern, tion of new units. Vancomycin, teicoplanin, and decaplanin are
because this confers resistance to a broad range of β-lactam all glycopeptides, but only vancomycin has been used in small
antibiotics. Carbapenemase-producing K. pneumoniae (KPC) animals.
are resistant not only to all β-lactam antibiotics, but also to fluo- Vancomycin was discovered in the 1950s, but has not been
roquinolones and aminoglycosides.12 K. pneumoniae that pro- frequently used in small animals because it is not absorbed
duce a plasmid-encoded metallo-β-lactamase enzyme (NDM-1) orally, so IV infusion is required for treatment of systemic
appeared in India in 2008 and have subsequently spread world- infections. When administered IV, it can be valuable for treat-
wide.13 The only options left for treatment of these infections ment of MDR gram-positive bacterial infections, such as
are tigecycline and polymyxins, both of which have been asso- methicillin-resistant staphylococcal infections, resistant entero-
ciated with emergence of resistance during treatment.14 The coccal infections, and encrusting cystitis caused by Corynebac-
potential for spread of K. pneumoniae isolates that are resistant terium urealyticum. The last is an emerging infection of dogs,
to all commercially available antibiotics now exists. Carapen- cats, and immunocompromised human patients, treatment of
emase (NDM-1) production was recently detected in an E. coli which requires surgical debridement of the bladder in conjunc-
isolate from a cat in the United States. tion with glycopeptide administration.15,16 Vancomycin is a
72 SECTION 2  Antiinfective Therapy

valuable agent for treatment of methicillin-resistant staphylo- Nucleic Acid Inhibitors


cocci, because resistance among these organisms is extremely
rare, and the bactericidal activity can be an advantage when Fluoroquinolones
treating immunocompromised patients. In human patients, oral Classification and Mechanism of Action
vancomycin has been used to treat gastrointestinal infections The fluoroquinolones bind to DNA gyrase (also known as topoi-
caused by resistant Clostridium difficile. somerase II) and topoisomerase IV, enzymes that cleave DNA
Vancomycin-resistant methicillin-resistant Staphylococcus during DNA replication. The result is disruption of bacterial
aureus (MRSA) isolates are extremely rare in people and have DNA and protein synthesis. For the veterinary fluoroquinolones
only been described in specific geographic locations. Vancomycin- enrofloxacin, orbifloxacin, and marbofloxacin, DNA gyrase is
resistant Staphylococcus pseudintermedius isolates have not yet the primary target for gram-negative bacteria and topoisomer-
been described. Vancomycin-resistant enterococci (VRE) are ase IV is the primary target for gram-positive bacteria. Because
well documented in human patients and can present a therapeutic topoisomerase IV has a lower affinity than DNA gyrase for this
challenge. Although currently rare, the existence of VRE has group of drugs, higher MICs are observed for gram-positive
been reported in companion animals.17 Resistance to vancomycin bacteria compared to the Enterobacteriaceae.
results from bacterial alteration of the terminal amino acid to Fluoroquinolones were created by addition of a fluorine group
which vancomycin binds. In order to reduce inappropriate use to nalidixic acid, which improves its spectrum of activity. Exam-
of vancomycin, some laboratories do not report results for van- ples are enrofloxacin, ciprofloxacin, difloxacin, orbifloxacin, and
comycin susceptibility unless reporting is indicated on the basis marbofloxacin. Newer-generation fluoroquinolones inhibit both
of resistance in the remainder of the susceptibility panel. DNA gyrase and topoisomerase IV in gram-positive bacteria,
Teicoplanin has a longer half-life and is up to 100 times leading to enhanced activity for treatment of gram-positive and
more lipophilic than vancomycin. It is not commercially avail- anaerobic bacterial infections and reduced likelihood of selection
able in the United States but is used in Europe. Vancomycin for resistant mutants. Pradofloxacin, levofloxacin, moxifloxa-
and teicoplanin appear to be equally efficacious in human cin, and gatifloxacin are third-generation fluoroquinolones. The
patients.18 only antibiotic in this group approved for use in companion ani-
mals is pradofloxacin, which is approved for use in cats in the
Adverse Effects United States, and approved for both dogs and cats in European
Vancomycin is not commonly used to treat small animals, countries.
and so adverse reactions have not been well documented. The Resistance to fluoroquinolones results from DNA gyrase
most common reaction in human patients is the “red man syn- mutations, decreased bacterial permeability, and increased
drome” caused by histamine release after rapid infusion. This drug efflux.19 Susceptibility to one fluoroquinolone generally
can be prevented through the use of antihistamines and slower predicts susceptibility to others, with the exceptions of third-
infusion rates. Other adverse reactions are rare. Previously generation fluoroquinolones (such as pradofloxacin) and cipro-
cited nephrotoxicity from vancomycin may be exaggerated floxacin, which has higher in vitro activity against P. aeruginosa
because of its frequent co-administration with aminoglyco- than other fluoroquinolones.20,21 Limited use of fluoroquino-
sides. Current formulations are of better quality and purity and lones has been recommended by some human and veterinary
less likely to produce kidney injury. Vancomycin is given by medical authorities in order to minimize further development
IV infusion over 30 to 60 minutes through a central or periph- of resistance, but a clear association between prescription of
eral catheter (Table 8-6). Trough plasma concentrations can be fluoroquinolones and increased resistance has not been estab-
monitored and should be greater than 10 µg/mL for skin and lished in companion animals. Nevertheless, other “first line”
soft tissue infections, and 10 to 20 µg/mL for more compli- drugs are often available for most infections, and the valuable
cated infections. Intramuscular administration is painful and fluoroquinolones can be reserved for cases that cannot tolerate
irritates tissues. other drugs, or when resistance to other drugs is more likely.

TABLE 8-6
Suggested Vancomycin Dosages for Small Animals
CLSI Breakpoints
Drug Dose (mg/kg) Interval (hours) Species Route Comments (µg/mL)
Vancomycin* 15 6-8 D IV Reconstituted drug is added to 0.9% ≤4 Enterococcus and
12-15 8 C NaCl or 5% dextrose and admin- Staphylococcus
istered over 30 to 60 minutes.
It can also be administered in 5%
dextrose as a CRI, with a loading
dose of 3.5 mg/kg, and then
1.5 mg/kg/hr IV. Therapeutic
drug monitoring recommended.

C, Cats; CLSI, Clinical and Laboratory Standards Institute; CRI, continuous-rate infusion; D, dogs.
*The use of vancomycin should be reserved for gram-positive infections that are susceptible to vancomycin but resistant to all other reasonable
alternatives, on the basis of culture and susceptibility testing.
CHAPTER 8  Antibacterial Drugs 73

A sufficiently high dose should be used to minimize selection of by divalent and trivalent cation-containing medications (e.g.,
resistant isolates. antacids) and supplements (aluminum, calcium, iron, zinc).
Use of these should be avoided if animals are receiving fluo-
Clinical Use roquinolones by the oral route. Oral absorption of the human
Fluoroquinolones are most valuable to treat gram-negative infec- drug ciprofloxacin is lower and more variable in dogs than
tions, because there are often few oral medication alternatives in people, which necessitates administration of a higher dose.
for these infections. The fluoroquinolones are also effective for In one study, oral absorption of generic ciprofloxacin tablets in
infections caused by staphylococci, but staphylococci tend to have dogs was 58%, but the coefficient of variation was 45%.23 In
higher MICs for fluoroquinolones than gram-negative bacteria. cats, only 22% to 33% of orally administered ciprofloxacin is
The activity of fluoroquinolones against anaerobes is variable. absorbed. Most fluoroquinolones are highly concentrated in the
Pradofloxacin has the greatest activity, followed by marboflox- urine, so they are useful for treatment of UTIs. Enrofloxacin
acin and enrofloxacin.22 Fluoroquinolones are concentration- is metabolized to ciprofloxacin, which is subsequently excreted
dependent antibiotics, so they are best administered as a single in the urine. Approximately half of the administered dose of
high daily dose. Doses for fluoroquinolones are listed in Table 8-7. marbofloxacin and orbifloxacin is excreted as unchanged drug
Fluoroquinolones are usually well absorbed after oral admin- in the urine. In contrast, difloxacin is excreted primarily in bile,
istration. Poor absorption occurs when they are complexed and little drug enters the urine.24

TABLE 8-7
Suggested Fluoroquinolone Dosages for Small Animals
Dose Interval
Drug (mg/kg) (hours) Species Route Comments CLSI Breakpoints (µg/mL)
Enrofloxacin 5-20 24 D IV, IM, PO Interactions can occur with certain ≤0.5 susceptible infections.
5 24 C* IM, PO oral medications (see text). Use higher dosages (10-20
Reduce dose or increase dosage mg/kg) for organisms with
interval with renal failure. Dilute MICs of 1 or 2 (except in
in fluids (e.g., 1:10 dilution) and cats). A dose of 5 mg/kg
infuse IV over 30 minutes. may be sufficient for UTIs.
Ciprofloxacin 20-30 24 D, C PO See enrofloxacin. Oral ­absorption In humans, ≤1 susceptible
­hydrochloride 10 24 D, C IV may be limited (see text). infections. More active
against Pseudomonas
aeruginosa than other
fluoroquinolones
Marbofloxacin 2.75-5.5 24 D, C PO See enrofloxacin. ≤1 susceptible infections. Use
higher doses for bacteria
with higher MICs.
Orbifloxacin 2.5-7.5 24 D, C PO See enrofloxacin. Orbifloxacin See marbofloxacin.
suspension provides lower and
more variable plasma levels than
the tablets. A dose of 7.5 mg/kg
should be used.
Difloxacin 5-10 24 D PO Urine concentrations may not be ≤0.5 susceptible ­infections
­hydrochloride sufficient for treating UTIs.
Moxifloxacin 10 24 D, C PO Moxifloxacin has enhanced activi- See marbofloxacin.
ty against gram-­positive bacteria
and anaerobes. In humans, urine
concentrations are not sufficient
for treatment of UTIs.
Pradofloxacin 3† 24 D, C PO See moxifloxacin for spectrum of Not established
5-7.5‡ C activity. Food reduces bioavail-
ability. Appears effective for
treatment of UTIs.

C, Cats; CLSI, Clinical and Laboratory Standards Institute; D, dogs; MIC, minimum inhibitory concentration; UTI, urinary tract infection.
*The use of enrofloxacin to treat cats is controversial because of the potential for irreversible retinal toxicity and blindness. Do not exceed a dose
of 5 mg/kg in cats. The use of alternative fluoroquinolones such as marbofloxacin, orbifloxacin, or pradofloxacin should be considered for cats.
†Tablet formulation. The use of pradofloxacin at high doses in dogs has been associated with myelosuppression and is off-label in the United States,

but it is approved for both dogs and cats outside the United States at a dose of 3 mg/kg (tablets).
‡Oral suspension for cats. The dose of the oral suspension for cats is higher (5-7.5 mg/kg) owing to reduced bioavailability.
74 SECTION 2  Antiinfective Therapy

Fluoroquinolones have variable protein binding, but this does of fluoroquinolones during the period of rapid growth in dogs
not inhibit distribution to tissue fluids. Because of good lipophilic- (from about 1 to 2 months of age to 28 weeks) should be avoided
ity, these drugs penetrate the prostate and respiratory secretions. if possible. Cats are thought to be more resistant to development
They also penetrate the ear canal, but high doses may be required of cartilage toxicity. Pradofloxacin has been associated with
for treatment of bacteria with relatively high MIC values.25 Fluo- myelosuppression in dogs at doses greater than 10 mg/kg.
roquinolones can attain high intracellular concentrations and can Fluoroquinolones inhibit some cytochrome P450 enzymes,
be used to treat infections caused by intracellular pathogens such which decreases metabolism of other drugs. This occurs with
as Mycoplasma spp. and some Mycobacterium spp. theophylline in dogs; therefore, the dose of theophylline should
be adjusted if administered concurrently. Like cephalosporins,
Adverse Effects fluoroquinolones can cause false-positive results on test strips
Fluoroquinolones can occasionally produce gastrointestinal for urine glucose detection. Cutaneous photosensitization
signs in dogs and cats, such as decreased appetite and vomit- occurs in human patients but is rare in animals. Other adverse
ing. Rapid IV administration can cause systemic hypotension, effects reported in humans, but not in dogs and cats, include
tachycardia, and cutaneous erythema, possibly as a result of cardiac arrhythmias, tendinitis and tendon rupture, altered glu-
histamine release.26 Neurologic signs, including tremors, ataxia, cose metabolism, and hepatic dysfunction.34
and seizures, can occur in dogs and cats treated with high doses
of parenteral fluoroquinolones. Although not well understood, Metronidazole
this is thought to result from fluoroquinolone interference with Mechanism of Action and Spectrum of Activity
the binding of γ-aminobutyric acid to its receptors in the central Metronidazole is a bactericidal nitroimidazole drug that is used
nervous system (CNS).27 In general, fluoroquinolones should primarily to treat anaerobic bacterial infections (both gram-
be used cautiously in animals prone to seizures. Enrofloxacin negative and gram-positive), which includes those caused by
causes hallucinations in humans and so is not marketed or Bacteroides fragilis and Clostridium spp., and infections with
approved for treatment of humans. certain protozoa, such as Giardia spp. Metronidazole diffuses
Cats treated with high doses of enrofloxacin (generally >5 into bacterial cells as a prodrug and is activated in the cyto-
mg/kg/day), especially parenteral enrofloxacin, have developed plasm. Once within the cell, the nitro group of metronidazole
blindness resulting from acute retinal degeneration, manifested as preferentially accepts electrons from electron transport proteins
bilateral mydriasis with tapetal hyperreflectivity.28,29 This results such as ferredoxin. A short-lived nitroso free radical is thus gen-
from a functional defect in a fluoroquinolone transport protein erated that damages DNA. The intermediate compounds then
in the cat, with subsequent accumulation of photoreactive drug decompose into non-toxic, inactive end products.
in the retina.30 In general, the blindness is irreversible, but if it is Metronidazole has antioxidant properties that are believed
detected early and medication is discontinued, some resolution to result from its ability to scavenge reactive oxygen species and
may occur. Orbifloxacin also can produce retinal injury in cats, modulation of neutrophil activity.35 It is often used at low doses
but the dose needed for this reaction is much higher than doses to treat inflammatory bowel disease in dogs and cats,36 but it
used therapeutically. Studies of marbofloxacin (reported by the is not known if the benefit is from the antibacterial activity or
manufacturer) and pradofloxacin31 showed no evidence of retinal direct effects on inflammatory cells.
toxicity in cats, even with high doses, although there are reports Resistance to metronidazole is rare among anaerobes.37
to the FDA of blindness in cats associated with the use of mar- Mechanisms of resistance include reduced drug uptake and
bofloxacin. Cats with renal insufficiency or those treated with decreased reduction activity. Other proposed mechanisms
medications that increase plasma fluoroquinolone concentrations include increased efflux, drug inactivation, and accelerated
may be more likely to develop this adverse effect. DNA repair.
Because they inhibit proteoglycan synthesis and chelate mag-
nesium, fluoroquinolones can cause cartilage and joint toxic- Clinical Use
ity in young animals, although the clinical importance of this In dogs and cats, metronidazole is used to treat anaerobic bacte-
has been questioned.32,33 In general, prolonged (>7 days) use rial and protozoal infections in a variety of tissues. Suggested

TABLE 8-8
Suggested Metronidazole Dosages for Small Animals
Drug Dose (mg/kg) Interval (hours) Species Route Comments
Metronidazole 15 12 D PO Reduce dose by 50% in animals with hepatic
10-15 24 C dysfunction. An intravenous solution, metroni-
dazole hydrochloride, is available but is highly
acidic. It should be diluted in 100 mL of IV
crystalloids and neutralized with 5 mEq sodium
bicarbonate per 500 mg for a pH of 6-7. Di-
luted solutions are stable for 24 hours.
Metronidazole 25 12 C PO More palatable formulation for cats may be
benzoate available from some compounding pharmacies.

C, Cats; D, dogs.
CHAPTER 8  Antibacterial Drugs 75

doses are listed in Table 8-8. Metronidazole is rapidly and almost DNA-dependent RNA polymerase in bacteria. This leads to
completely absorbed from the gastrointestinal tract, diffuses impaired RNA synthesis. Only the semisynthetic drug rifampin
across lipid membranes, and achieves therapeutic concentrations (also known as rifampicin) has been used to any great extent in
in tissue fluids. Metronidazole is metabolized by the liver. The small animal medicine. Rifampin is active against staphylococci,
metabolites, along with intact drug, are excreted in the urine. streptococci, and only a few gram-negative organisms. When
used as a component of antimycobacterial treatment regimes, it
Adverse Effects increases the chance of treatment success. Rifabutin is effective
The main adverse effect of metronidazole is neurotoxicity, which for treatment of some mycobacterial infections that are resistant
tends to occur with high doses (>30 mg/kg/day) or in animals to rifampicin.43
with hepatic dysfunction. Clinical signs of neurotoxicity are It has been recommended that rifampin be used in combina-
reversible on discontinuation of the drug and include lethargy, tion with other antimicrobials to reduce the risk of emergence
truncal ataxia, hypermetria, intention tremors, head tilt, falling, of rifampin resistance, which occurs quickly when rifampin
vertical nystagmus, extensor rigidity, opisthotonos and seizures, is used alone. However, this applies primarily when it is used
and reflect damage to the vestibular system.38,39 Although not for long-term treatment, such as for mycobacterial infections.
fully understood, neurotoxicity is believed to result from antag- Monotherapy has been successful when rifampin has been used
onism of γ-aminobutyric acid in the cerebellar and vestibular to treat methicillin-resistant Staphylococcus infections, which
systems. Neurologic signs resolve more quickly when treated accounts for most of the current small animal use. If combina-
with a low dose of diazepam (0.4 mg/kg PO q8h for 3 days).38 tion treatment is pursued for treatment of methicillin-resistant
Bilateral symmetrical cerebellar lesions, most commonly in the staphylococci, there are often few effective alternatives with
dentate nucleus have been reported on magnetic resonance which to combine rifampin, because these infections are typi-
imaging in human patients with metronidazole toxicity,40 and cally multidrug resistant. Resistance to rifampin results from a
multifocal necrotic foci were detected at necropsy in the brain- single mutation that leads to an altered RNA polymerase that
stem of a cat.39 Administration of metronidazole to animals does not effectively bind rifampin.
with a history of metronidazole toxicosis is not recommended.38
Less commonly, metronidazole causes decreased appetite, Clinical Use
vomiting, and diarrhea. Metronidazole suspension has a bitter One of the major advantages of rifampin is its high degree of
taste that may be unpalatable to cats, which can be overcome lipid solubility, which provides the degree of intracellular pen-
by administration of the drug in gel capsules. Although not etration required for treatment of infections caused by intracel-
approved by the FDA, metronidazole benzoate is a more pal- lular bacteria such as Mycobacterium spp. and Brucella canis
atable formulation for cats and pharmacokinetic studies have infections. Rifampin has been used to treat bartonellosis in com-
demonstrated good absorption.41 There have been concerns bination with doxycycline.44,45 Because of its activity against
that relate to toxicity from the benzoic acid component of the staphylococci and the fact that it can be administered orally, it
formulation, which can cause CNS signs, blindness, and respi- has been used for treatment of methicillin-resistant staphylococ-
ratory problems at high doses. However, this possibility is con- cal infections in small animals. For other infections, rifampin is
sidered to be unlikely.41 Because metronidazole benzoate is only less attractive because of its adverse effect profile.
62% metronidazole, dosage adjustment is required when this Rifampin dosing is shown in Table 8-9. Rifampin is rapidly
formulation is used. Metronidazole inhibits hepatic microsomal absorbed from the gastrointestinal tract and metabolized by
enzymes and so may increase the concentration of drugs such as the liver. Active metabolites are excreted in bile and to a lesser
warfarin and cyclosporine. It also lowers serum lipid concentra- extent the urine.
tions (including cholesterol concentrations) in human patients.42
Adverse Effects
Rifamycins Vomiting and anorexia are common in dogs treated with
Mechanism of Action and Spectrum of Activity rifampin. The drug can impart a red-orange color to the urine
Named after the French crime movie Rififi, rifamycins are and, to a lesser extent, tears, saliva, the sclera, and mucous
bactericidal antimicrobials that inhibit the β-subunit of membranes. Rifampin can also be associated with increased

TABLE 8-9
Suggested Rifampin Dose for Small Animals
Interval CLSI Breakpoints
Drug Dose (mg/kg) (hours) Species Route Comments (µg/mL)
Rifampin 5 12 D, C PO Preferably use in combination with other ≤1 for susceptible
drugs. Avoid in animals with hepatic organisms (usual-
dysfunction. Do not administer with fatty ly MICs of gram-
meals. Drug interactions may occur (see positive bacteria
text). Monitor liver enzymes with prolonged fall below this
treatment. May impart a red-orange color breakpoint)
to the urine and tears.

C, Cats; CLSI, Clinical and Laboratory Standards Institute; D, dogs; MIC, minimum inhibitory concentration.
76 SECTION 2  Antiinfective Therapy

serum liver enzyme activities and hepatopathy. Doses that Clinical Use
exceed 10 mg/kg per day in dogs increase the risk of liver injury. TMS antibiotics have a broad spectrum of activity and can be
In humans, fever, cutaneous reactions, thrombocytopenia, acute used to treat gram-positive and many gram-negative bacterial
intravascular hemolytic anemia, and acute tubulointerstitial infections, as well as some protozoal infections. Enterococci
nephritis have been reported, which appear to have an immune- are intrinsically resistant to TMS antibiotics. Many anaerobic
mediated pathogenesis.46 Rarely, pancreatitis has been reported bacteria are not treated successfully because despite in  vitro
in human patients receiving rifampin. Adverse effects have not susceptibility, the drugs are less active in tissues where anaer-
been reported in cats, but some of these risks may apply to cats obes proliferate. TMS antibiotics are the treatment of choice for
as well as dogs. Because of poor palatability, it is a challenge to nocardiosis. They have also been used successfully for preven-
administer to cats. tion of bacterial infections in dogs being treated with myelosup-
Rifampin is one of the most potent inducers of hepatic pressive chemotherapeutics such as doxorubicin.49 Prophylactic
microsomal enzymes and efflux proteins, such as P-glycoprotein. protocols have selected for resistant bacteria in humans,50 but
Subsequently, it can increase the clearance of several co-­ this has not been reported in animals. TMS combinations are
administered drugs (thereby decreasing their efficacy), such available as either trimethoprim-sulfadiazine or trimethoprim-
as glucocorticoids, cyclosporin, itraconazole, and digoxin. In sulfamethoxazole (the latter is a human medical preparation),
human patients, recovery from the effect of rifampin on hepatic which are used interchangeably. However, the veterinary
microsomal enzymes can take 4 weeks.47 formulation of trimethoprim-sulfadiazine is rarely available
commercially, and most veterinarians rely on trimethoprim-
Trimethoprim-Sulfonamides sulfamethoxazole. No difference in activity between the two
Mechanism of Action formulations has been documented, but this has never been eval-
Trimethoprim and sulfonamides act synergistically to inhibit uated. The primary difference between the formulations is that
folic acid metabolism by bacteria. The combination interferes sulfadiazine is more water soluble and is excreted in the urine in
with purine and therefore DNA synthesis. Trimethoprim inhib- an unchanged form. A similar formulation to the trimethoprim-
its bacterial dihydrofolate reductase. Sulfonamides are chemical sulfonamides for use in veterinary medicine is ormetoprim sul-
analogs of para-aminobenzoic acid (PABA), and competitively fadimethoxine, which has been used for the same indications as
inhibit the incorporation of PABA into dihydropteroic acid by trimethoprim-sulfonamides. Doses for TMS combinations are
the enzyme dihydropteroate synthetase, which also leads to listed in Table 8-10.
decreased folic acid synthesis. When administered separately, TMS is well absorbed after oral administration. As a weak
each drug is bacteriostatic, but the combination is bactericidal. base, trimethoprim is able to penetrate the blood-prostate bar-
Preparations used to treat dogs and cats include a 1:5 ratio of rier and can be used to treat prostatic infections. Both trim-
trimethoprim to sulfonamide. Because folic acid is of dietary ethoprim and sulfonamides are metabolized to some extent by
origin in animals, only bacterial purine synthesis is affected. the liver. Both active drug and inactive metabolites appear in
Resistance to trimethoprim-sulfonamide (TMS) antibiotics the urine. Active TMS is highly concentrated in urine and is an
most commonly results from plasmid-mediated production of appropriate first choice for treatment of UTIs in dogs and cats.51
altered dihydrofolate reductase or dihydropteroate synthetase,
with reduced binding affinities. Other mechanisms include over- Adverse Effects
production of dihydrofolate reductase or PABA by bacteria, TMS antimicrobials cause a wide range of adverse effects, which
and reduced bacterial permeability to trimethoprim and sulfon- have somewhat limited their use. These reactions are primar-
amides. Multiple mechanisms may operate simultaneously.48 ily caused by the sulfonamide component. Some of the adverse

TABLE 8-10
Suggested Trimethoprim-Sulfonamide Dosages for Small Animals
Dose Interval CLSI Breakpoints
Drug (mg/kg) (hours) Species Route Comments (µg/mL)
Trimethoprim- 30* 12 D, C PO, IV Avoid in animals with hepatic ≤2/38 for suscep-
sulfamethoxazole, failure. Reduce dose in animals tible infections
trimethoprim-­ with renal insufficiency. Obtain
sulfadiazine baseline Schirmer tear test and
monitor this and the CBC with
prolonged treatment. Each 5-mL
vial of the injectable preparation
should be diluted in 75-125 mL
of 5% dextrose and adminis-
tered IV over 1 hour.
Ormetoprim- 55 on first day, 24 D PO See trimethoprim-­ Use information
sulfadimethoxine and then 27.5* sulfamethoxazole. for TMS.

C, Cats; CLSI, Clinical and Laboratory Standards Institute; D, dogs; TMS, trimethoprim-sulfonamide.
*Dose listed is for combined components, (e.g., 5 mg of trimethoprim and 25 mg of sulfonamide).
CHAPTER 8  Antibacterial Drugs 77

effects may be attributed to the inability of dogs to acetylate gram-negative bacteria than gram-positive bacteria because of a
sulfonamide compounds. People who are “slow acetylators” are greater presence of an outer membrane in gram-negative organ-
also more prone to adverse effects from sulfonamides. isms. The end result is disrupted cell permeability. Subsequently,
Gastrointestinal signs such as vomiting can occur. Other the bacterial cell takes up the aminoglycoside molecules, and
common adverse effects in dogs are hypersensitivity reactions they become trapped irreversibly within the cytoplasm. Because
and include keratoconjunctivitis sicca, fever, polyarthritis, this is an oxygen-dependent process, anaerobes are intrinsically
cutaneous drug eruptions,52,53 immune-mediated thrombocy- resistant to aminoglycosides. Once within the cell, aminoglyco-
topenia, hemolytic anemia, hepatitis, pancreatitis, meningitis, sides bind to the 30S subunit of the bacterial ribosome, which
interstitial nephritis, glomerulonephritis, and aplastic anemia­ results in decreased protein synthesis.
(Figure 8-3).54-57 These events are relatively uncommon, but Aminoglycosides, particularly gentamicin, amikacin, and
have been documented well enough in dogs that it has discour- tobramycin, are highly active compounds for which resistance
aged their routine use among some veterinary clinicians. Revers- is uncommon. The most common mechanism of aminoglyco-
ible hypothyroidism can occur after periods of treatment as side resistance is enzymatic modification of the drug by bacte-
short as 10 days.58 Hepatic necrosis can occur as an idiosyncratic ria. More than 50 modifying enzymes have been identified.62
reaction.59 Ormetoprim has been associated with neurologic Reduced drug uptake by bacteria may also occur. Enterococci
signs in dogs, including tremors, anxiety, and seizures. Schirmer are intrinsically resistant to low levels (4 to 250 µg/mL) of ami-
tear tests should be performed before sulfonamides are admin- noglycosides because of their anaerobic metabolism, and thus
istered to dogs, tear tests should be monitored when prolonged they require testing for high-level resistance using higher concen-
treatment is required (e.g., greater than 1 week), and the drug trations of aminoglycosides. Resistance to one aminoglycoside
should be avoided in dogs with keratoconjunctivitis sicca. Sul- does not imply resistance to others. For example, tobramycin
fonamide cystolithiasis can occur in dogs treated with high doses and amikacin are consistently more active than gentamicin, and
of sulfonamides.60 Doberman pinschers may be more susceptible kanamycin is one of the least active aminoglycosides. Because of
than other breeds to adverse effects of TMS because of their these differences, a susceptibility panel should include gentami-
inability to detoxify certain sulfonamide metabolites; in another cin and either amikacin or tobramycin.
study, Samoyeds and miniature schnauzers also appeared to be
more susceptible.55,61 Clinical Use
Aminoglycosides are primarily used to treat gram-negative
Protein Synthesis Inhibitors bacterial infections and MDR staphylococcal infections. They
should not be used to treat anaerobes, and Streptococcus species
Aminoglycosides are better treated with β-lactams. Aminoglycosides have usu-
Mechanism of Action ally been administered together with cell wall–active antimicro-
All aminoglycosides contain an essential six-membered ring with bial drugs (β-lactams and glycopeptides), but this is no longer
amino-group substituents and are highly positively charged. believed to be a therapeutic advantage. They are very water sol-
Aminoglycosides with names that end in -mycin derive directly uble and poorly lipid soluble, so they penetrate tissue fluids well,
or indirectly from Streptomyces spp. (e.g., neomycin). Those but do not readily penetrate tissues that are dependent on lipid
with names ending in -micin are derived from Micromonospora diffusion (prostate, brain, eye, or cerebrospinal fluid). Penetra-
(e.g., gentamicin). tion of bile is also poor. Oral absorption is negligible, and they
Aminoglycosides have multiple mechanisms of action. They should not be administered orally unless specifically intended
bind electrostatically to the bacterial outer membrane and dis- to treat intestinal bacteria (e.g., oral neomycin for treatment of
place cell wall Mg2+ and Ca2+, which normally link adjacent hepatic encephalopathy). Doses are shown in Table 8-11. Ami-
lipopolysaccharide molecules. This initial binding is greater in noglycosides also may be administered topically. Because they
penetrate bronchial secretions poorly, nebulization has been
used in order to overcome this problem in human patients as
well as dogs with resistant gram-negative bacterial broncho-
pneumonia (see Table 8-11).63 Aminoglycosides are excreted
unchanged by the kidney and concentrate 25- to 100-fold in
urine within an hour of drug administration, so they can be used
to treat resistant gram-negative bacterial UTIs. They also accu-
mulate in renal tissue and remain active for several days after
dosing is discontinued. Because they are not well distributed
into fat, dose reduction may be indicated for obese animals.
Despite their primary mechanism of action being protein
synthesis inhibition, aminoglycosides are rapidly bactericidal
with a prolonged, concentration-dependent postantibiotic
effect. Transient high peak drug concentrations result in faster
killing and greater bactericidal activity than lower concentra-
tions over a prolonged period of time (i.e., they are concentra-
FIGURE 8-3  Right eye of a 7-year-old intact male golden retriever that developed ker- tion-dependent antimicrobial drugs). Once-daily dosing of a
atoconjunctivitis sicca after administration of trimethoprim-sulfamethoxazole for 3 weeks high dose of an aminoglycoside is therefore preferred because
for septic peritonitis. The owner noticed the dog pawing at his eyes. Mucoid ocular discharge it achieves optimal bacterial killing but minimizes toxicity. In
is present at the medial canthus. Schirmer tear test strip results were 0 mm for both eyes. order to further minimize toxicity, aminoglycosides are gener-
(Image courtesy University of California, Davis Veterinary Ophthalmology service.) ally infused slowly. In human patients, it is recommended they
78 SECTION 2  Antiinfective Therapy

TABLE 8-11
Suggested Aminoglycoside Dosages for Small Animals
Dose Interval CLSI Breakpoints
Drug (mg/kg) (hours)* Species Route Comments (µg/mL)
Gentamicin 9-14 24 D IV, IM, SC Ensure adequate fluid and electrolyte ≤2 susceptible
sulfate 5-8 24 C balance during treatment. Avoid infections
in animals with renal impairment.
Monitor BUN and creatinine.
Amikacin and tobramycin are more
active against some gram-negative
bacteria than gentamicin, but
gentamicin is preferred for Serratia
infections. Do not mix in vials with
other drugs.
Amikacin 15-30 24 D IV, IM, SC See gentamicin. ≤16 susceptible
10-14 24 C infections
Tobramycin 9-14 24 D IV, IM See gentamicin. For nebulization, 160 ≤4 susceptible
sulfate 5-8 24 C mg of nebulization solution† q12h infections
for 28 days. Preferred aminoglyco-
side for Pseudomonas infections.
Kanamycin 20 24 D, C IV or IM See gentamicin. Less active than genta- ≤16 susceptible
sulfate micin and amikacin. infections
Neomycin 10-20 6-12 D, C PO Not absorbed systemically; used to Not applicable
treat hepatic encephalopathy. Use
cautiously in animals with renal
disease and avoid use for >14 days.

C, Cats; CLSI, Clinical and Laboratory Standards Institute; D, dogs; MIC, minimum inhibitory concentration.
*Once-daily administration is preferred except for enterococcal endocarditis, where the continuous presence of both an aminoglycoside and a penicil-
lin is recommended (see Chapter 86).
†TOBI nebulization solution. Injectable tobramycin or gentamicin can be used but may be associated with bronchospasm. They should be diluted in

3 mL of sterile saline, and albuterol should be administered just before nebulization.

be administered over a 15- to 30-minute period. Subcutaneous the cytoplasm, where interference with mitochondrial function
and intramuscular injections of water-soluble formulations of occurs. Aminoglycosides also bind membrane phospholipids
aminoglycosides are also acceptable routes of administration, and alter their turnover. Damaged renal tubular epithelial cells
although they may cause pain in some animals. are shed into and obstruct the tubular lumen, which leads to a
build-up of pressure within the glomerulus, leading to decreased
Adverse Effects glomerular filtration rate. Disrupted proximal tubular cell func-
Aminoglycosides have the potential to cause nephrotoxicity, tion leads to delivery of excessive water and electrolytes to
ototoxicity, and neuromuscular blockade. Their cationic struc- the distal nephron, which in turn triggers tubuloglomerular
ture and the high anionic phospholipid concentrations in the feedback, with constriction of afferent arterioles and further
kidney and inner ear play an important role in toxicity. In the decreases in the glomerular filtration rate.65 Tubuloglomerular
presence of gastrointestinal ulceration, systemic absorption of feedback is designed to protect the body from massive loss of
orally administered aminoglycosides can occur, with associated water and electrolytes.
nephrotoxicity or ototoxicity.64 Risk factors for nephrotoxicity include older age, reduced
Nephrotoxicity results from apoptosis and necrosis of renal renal function, concomitant liver disease, dehydration, sodium
tubular epithelial cells, and direct damage to the glomerulus as depletion, hypokalemia, and concurrently administered neph-
well. The mechanisms involved are complex.65 Recovery can rotoxic drugs, most commonly nonsteroidal antiinflammatory
occur when the aminoglycoside is removed, but because ami- drugs but also furosemide and cyclosporin. Although genta-
noglycosides accumulate in renal tissue, the insult can persist micin nephrotoxicity can occur in young animals,66 young age
for some time even after treatment is discontinued. Efforts to is not a contraindication to use of aminoglycosides, which are
understand the mechanism of nephrotoxicity should lead to used widely to treat human neonatal sepsis.67 The chance of
improved preventative measures and increased applications for toxicity increases with more prolonged treatment periods (≥3
aminoglycoside antibiotics. Aminoglycosides bind to megalin- days). Gentamicin and amikacin are more toxic than tobramy-
cubulin complexes on the surface of the cell and are endocy- cin. Volume and salt loading do not decrease toxicity, but high
tosed into the cytoplasm, where they accumulate in lysosomes. levels of dietary calcium and protein have protective effects.68
The lysosomes break down, with resultant drug leakage into The best preventative measure is to avoid these drugs in any
CHAPTER 8  Antibacterial Drugs 79

TABLE 8-12
Suggested Chloramphenicol Dosages for Small Animals
Dose Interval CLSI Breakpoints
Drug (mg/kg) (hours) Species Route Comments (µg/mL)
Chloramphenicol, 40-50 6-8 D PO, IV, IM Avoid long-term use in cats. ≤4 for streptococci,
chloramphenicol 12.5-20 12 C Monitor CBC with long- ≤8 for other sus-
sodium succinate term use. Warn owners ceptible bacteria
that human exposure to
chloramphenicol may cause
bone marrow disease. Drug
interactions may occur (see
text). Avoid in animals
with hepatic failure.
Florfenicol 20 6 D, C PO, IM Not approved for use in See chlorampheni-
22 8 dogs and cats. Doses and col.
adverse effects have not
been well studied.

C, Cats; CLSI, Clinical and Laboratory Standards Institute; D, dogs.

patients with preexisting renal disease. If renal disease develops followed by two additional time points in order to estimate the
after initiating treatment, the drug should be substituted with a slope of the elimination curve. Trough levels could also be per-
safer drug. formed to minimize the chance of toxicity, with the recommen-
Ototoxicity results from damage to the cochlear or the ves- dation that if high trough levels are detected, the drug should
tibular apparatus and is more likely to occur in animals with be discontinued. In patients with normal renal function, trough
renal impairment or that are dehydrated. Cochlear toxicity levels with once-daily dosing should be in the “undetectable”
results from damage to the hair cells of the organ of Corti, range.
whereas vestibular toxicity results from damage to hair cells at
the tip of the ampullae cristae. Auditory toxicity may be more Chloramphenicol
common in dogs, whereas cats tend to develop vestibular toxic- Mechanism of Action and Spectrum of Activity
ity.69 Cochlear toxicity in dogs and cats may be underrecog- Chloramphenicol binds to the 50S subunit of the bacterial
nized, because it may be overlooked by pet owners. In human ribosome and inhibits bacterial protein synthesis. It is usually
patients, signs of vestibular toxicity can disappear with time described as bacteriostatic, but there is some evidence that
because of compensatory mechanisms. Ototoxicity is generally chloramphenicol may be more bactericidal than previously
irreversible, but in some human patients, gradual improvement thought. Like other ribosomal inhibitors, chloramphenicol
in hearing can occur.70 has a broad spectrum of activity, which includes gram-positive
Neuromuscular blockade is a rare but potentially life-­ and gram-negative bacteria, anaerobes, and some rickettsial
threatening complication of aminoglycoside treatment. Severe pathogens. Because chloramphenicol can cause aplastic anemia
neuromuscular blockade has not been reported in cats and dogs, in humans, its use in humans has greatly diminished, and it is
but experimental studies show it does occur when aminoglyco- only used for the treatment of MDR bacterial infections where
sides are used in conjunction with some anesthetic agents.71-73 few or no other antimicrobial drugs are useful. Thiamphenicol
Neuromuscular blockade results from decreased release of and florfenicol are related compounds, the availability of which
acetylcholine at neuromuscular junctions, and decreased sensi- varies from country to country. Although thiamphenicol was
tivity of the postsynaptic neuron to acetylcholine. Autonomic initially promoted as not inducing aplastic anemia in humans,
blockade can also occur. The use of aminoglycosides should be cases of aplastic anemia have now been identified.76 Florfenicol
avoided in animals with neuromuscular disease such as myas- is more active than chloramphenicol and thiamphenicol, but has
thenia gravis, because aminoglycosides have the potential to a shorter half-life in dogs and thus requires more frequent dos-
exacerbate the clinical signs.74 ing (e.g., more often than every 8 hours) in this species. It was
Measurement of serum aminoglycoside concentrations can developed for food animal species where use of chlorampheni-
be used to ensure adequate plasma concentrations or to mini- col is illegal.
mize toxicity.75 Assays are offered by specialized veterinary Resistance to chloramphenicols results from porin mutations,
pharmacology laboratories or through human hospitals. Ami- drug efflux, or production of chloramphenicol acetyltransferase
noglycoside doses that result in peak plasma concentrations enzymes, which inactivate the antibiotic.77 Resistance to one
approximately 8 to 10 times the MIC produce the optimal bac- chloramphenicol derivative predicts resistance to the others.
tericidal effect. The effective volume of distribution for amino-
glycosides is often higher in septic patients or animals that have Clinical Use
a “third space” problem (such as pleural effusion), which may Doses are shown in Table 8-12. Chloramphenicol is absorbed
necessitate a higher dose. To perform therapeutic drug monitor- orally with or without food. Tablets and capsules have simi-
ing, the optimum time points are 1 hour after injection (Cmax), lar oral absorption, but the extent of oral absorption varies
80 SECTION 2  Antiinfective Therapy

considerably among dogs. Chloramphenicol distributes well to macrolide antibiotics, such as clarithromycin and roxithromy-
extracellular fluids of most tissues. Because of its high degree cin, have been referred to as “advanced-generation” macrolides.
of lipid solubility, it also diffuses to tissues with barriers, such These have an extended spectrum of activity, with greater effi-
as the CNS and the eye. Topical ophthalmic preparations cacy against atypical and some gram-negative bacteria, a longer
achieve high concentration in the aqueous humor. In dogs, most duration of antimicrobial activity, … improved gastrointestinal
absorbed chloramphenicol is metabolized by glucuronidation in absorption, greater intracellular concentrations, and a marked
the liver, with inactive metabolites excreted by the kidney. The reduction in gastrointestinal adverse effects. Clarithromycin has
use of chloramphenicol should therefore be avoided in animals improved activity against streptococci and staphylococci com-
with hepatic failure. Provided renal failure is not present, suf- pared with erythromycin.
ficient active drug is excreted in the urine to enable treatment Azithromycin is an azalide, because unlike other macrolides,
of UTIs that are resistant to other drugs, such as methicillin- it contains nitrogen. Azithromycin is extensively concentrated
resistant staphylococci and resistant enterococci. within cells and is retained in tissues for prolonged periods, with
a half-life of 30 hours in the dog and 35 hours in the cat. In
Adverse Effects human patients, a 5-day course provides therapeutic tissue con-
Exposure of humans to even small amounts of chloramphenicol centrations for at least 10 days. Although azithromycin has an
has the potential to lead to irreversible aplastic anemia, an idio- improved spectrum against gram-negative bacteria, it has less
syncratic reaction, weeks to months after discontinuing treat- activity against gram-positive bacteria than erythromycin.82,83
ment. The reaction is rare (1 in 24,000 to 1 in 40,000 exposed Susceptibility to erythromycin often predicts susceptibility
humans) and most commonly occurs when human patients are to other macrolide antimicrobial drugs. Resistance to macro-
treated with oral formulations.78 Whether use of ophthalmic lides and lincosamides results from decreased bacterial per-
preparations in human patients poses a risk has been contro- meability (for gram-negative bacteria), alteration in the target
versial.79 Nevertheless, it is recommended that owners of pets site, increased drug efflux, and enzymatic inactivation of cer-
that are being treated with chloramphenicol wear gloves when tain macrolides by bacterial esterases. Alteration in the target
handling the drug and to avoid splitting tablets at home. Chlor- site involves production of a ribosomal methylase that adds a
amphenicol commonly causes gastrointestinal adverse effects in methyl group to the 50S subunit RNA, preventing the macro-
dogs and cats, including anorexia, hypersalivation, and vomit- lide from binding to the ribosome. Expression of the methylase
ing. High doses or prolonged treatment can result in reversible enzyme confers high-level resistance to azithromycin, clarithro-
bone marrow suppression, which usually resolves within days mycin, and also clindamycin.
after discontinuation of treatment. High doses and prolonged
treatment are needed to produce this effect in dogs, but cats are Clinical Use
more susceptible and can be at risk within 2 weeks of adminis- Macrolides and lincosamides are primarily used to treat gram-
tration. Chloramphenicol is a potent inhibitor of P450 enzymes, positive bacterial infections. They are generally considered to
and so the potential for significant drug interactions exists. Flor­ be bacteriostatic. Doses are shown in Table 8-13. Erythromycin
fenicol has not been widely used in dogs and cats, and adverse tablets have a protective coating to prevent degradation by gas-
effects have not yet been reported. tric acid. After absorption, erythromycin is distributed well to
most tissues and body fluids, with the exception of the CNS and
Macrolides and Lincosamides urine. It is excreted in high concentrations in bile, followed by
Classification and Mechanism of Action enterohepatic circulation, and ultimately eliminated in the feces.
The macrolides and lincosamides (clindamycin and lincomycin) Non-absorbed drug is also present in the feces. It has activity
are not chemically related but possess similar mechanisms of against some anaerobes, spiral bacteria (particularly Campylo-
action, resistance, and antimicrobial activity. Macrolides and lin- bacter), and many intracellular bacteria. Tylosin has been used
cosamides inhibit protein synthesis by binding to the 50S subunit to treat chronic colitis in dogs84 and is also active against Cam-
of bacterial ribosomes. Because they are weak bases, they concen- pylobacter and enteric Clostridium spp.
trate in the relatively acidic interior of leukocytes. The high con- Clarithromycin and azithromycin have considerably longer
centrations in leukocytes may also produce immunomodulatory half-lives than erythromycin. The intracellular concentration of
effects. Macrolides, particularly the newer agents such as azithro- clarithromycin is about two- to sixfold that in plasma. Clar-
mycin, can impair leukocyte function, interfere with superoxide ithromycin is preferred over erythromycin or azithromycin for
production by mast cells, suppress proinflammatory cytokine treatment of rapidly growing mycobacterial infections in dogs
release, and produce apoptosis.80,81 These effects may suppress and cats (usually in combination with rifampin) (see Chapter
inflammatory reactions in some tissues, but the significance of 44). In humans, clarithromycin is extensively metabolized by
these effects has not been explored in small animal infections. the liver to a variety of specific active and inactive metabo-
The first macrolide antibiotic to be discovered was eryth- lites, with some excretion of active drug and its metabolites
romycin, a derivative of Streptomyces erythreus. Despite the into urine. Formation and excretion of these metabolites have
common occurrence of gastrointestinal adverse effects, poor not been well studied in dogs and cats. Azithromycin is largely
gastrointestinal absorption, and an extremely short half-life, excreted unchanged in bile.
erythromycin continues to have application in human medicine Because of its prolonged tissue retention, serum concentra-
because of its safety in pregnancy and utility in human patients tions of azithromycin do not reflect tissue concentrations, and
who are allergic to penicillin. Because these concerns are less so the antibiotic can still be effective even if the serum con-
important in dogs and cats, erythromycin is less commonly used centration is less than the MIC. Intracellular concentrations
in small animal patients. of azithromycin are 10- to 100-fold those in serum, concen-
Since the discovery of erythromycin, several semisynthetic trations that are considered bactericidal. Concentrations can
macrolides have become available. Azithromycin and other new remain high within tissues several days after the drug has been
CHAPTER 8  Antibacterial Drugs 81

TABLE 8-13
Suggested Macrolide and Lincosamide Dosages for Small Animals
Dose Interval CLSI Breakpoints
Drug (mg/kg) (hours) Species Route Comments (µg/mL)
Erythromycin 10-20 8 D, C PO Do not administer to rabbits ≤0.25 for streptococci and
or rodents, as this may cause ≤0.5 for other suscep-
fatal diarrhea. Drug interac- tible organisms
tions may occur (see text).
Tylosin 7-15 12-24 D, C PO See erythromycin. For colitis in
8-11 12 IM dogs, use 20 mg/kg q8h with
food, and taper to q24h if a
response occurs.*
Clarithromycin 7.5 12 D, C PO Drug interactions may occur ≤2 for staphylococci.
(see text). Consider dosage Respiratory cures may
reduction in severe renal be possible for organ-
failure. isms with MICs up to 8,
because drug concen-
trates within the lung.
Azithromycin 5-10 24 D, C PO ≤ 2 for susceptible bacte-
ria. Serum MIC values
may not predict tissue
concentrations.
Lincomycin 15-25 12 D, C PO See erythromycin. A dose of 0.25 for streptococci and
­hydrochloride 10 mg/kg PO q12h has been ≤ 0.5 for other suscep-
suggested for pyoderma. tible organisms
Clindamycin 11-33 12 D PO See erythromycin. For IV See lincomycin.
­hydrochloride, 11 24 C PO administration, dilute 1:10 in
clindamycin 10 12 D, C IV, IM 0.9% saline and administer
­phosphate over 30-60 minutes. The oral
suspension may be unpalat-
able to cats. For toxoplasmo-
sis, use 25 mg/kg q12h PO.

C, Cats; CLSI, Clinical and Laboratory Standards Institute; D, dogs; MIC, minimum inhibitory concentration.
*20 mg/kg is equal to 1/8th teaspoon of tylosin phosphate (Tylan) for a 20-kg dog.

discontinued. Despite the improved spectrum of activity of injection). Clindamycin is also used to treat anaerobic bacterial
clarithromycin and azithromycin, with activity against some infections, as well as toxoplasmosis and neosporosis (see Chap-
gram-negative bacteria and intracellular pathogens, many of ters 72 and 73).
these organisms are resistant. The primary indication for these
antibiotics in small animals is in combination with other drugs Adverse Effects
such as rifampin for treatment of mycobacteriosis or bartonel- Although otherwise very safe, erythromycin frequently causes
losis, or atovaquone for treatment of certain systemic protozoal vomiting, decreased appetite and nausea because of stimu-
infections such as babesiosis and cytauxzoonosis. Azithromycin lation of receptors of the gastrointestinal hormone motilin,
has been used to treat secondary bacterial infections in cats with which increases gastrointestinal smooth muscle activity.86
upper respiratory tract disease because its long half-life permits Like other poorly absorbed oral antimicrobials, it can pro-
convenient dosing on an every-48- or every-72-hour basis. duce diarrhea due to changes in the gastrointestinal flora.
Despite its popularity for treating upper respiratory infections Although diarrhea and pseudomembranous colitis are seri-
in cats, one study showed no benefit to this protocol over use of ous complications of clindamycin treatment in people, these
amoxicillin,85 and the potential for overuse of azithromycin in problems are uncommon in small animals. Adverse effects are
this setting raises concerns in regard to selection for resistance rarely reported for clarithromycin and azithromycin. Eryth-
to azithromycin. romycin and azithromycin inhibit P450 enzymes and so can
Lincomycin is active against gram-positive bacteria and has increase the concentration of other drugs that are adminis-
been used to treat staphylococcal pyoderma. However, the tered concurrently.
use of lincomycin has largely been replaced by clindamycin, Clindamycin and lincomycin can cause vomiting and diar-
which has similar activity, favorable pharmacokinetics, and rhea, especially when clindamycin is used at high doses. Oral
widely available formulations (tablets, capsules, oral liquid, and clindamycin hydrochloride can cause esophagitis in cats.87,88
82 SECTION 2  Antiinfective Therapy

TABLE 8-14
Suggested Linezolid Dose for Small Animals
CLSI Breakpoints
Drug Dose (mg/kg) Interval (hours) Species Route Comments (µg/mL)
Linezolid* 10 8 D PO, IV If administered with other ≤4 for staphylococ-
drugs IV, the adminis- ci, ≤2 for Entero-
tration line should be coccus spp.
flushed first.

CLSI, Clinical and Laboratory Standards Institute; D, dogs.


*The use of linezolid should be reserved for gram-positive infections that are susceptible to linezolid but resistant to all other reasonable alterna-
tives, on the basis of culture and susceptibility testing.

Oxazolidinones (Linezolid) tetracycline, oxytetracycline, and the semisynthetic tetracycline


Mechanism of Action and Spectrum of Activity doxycycline. Doxycycline and minocycline are longer-acting tet-
Linezolid is a synthetic antibiotic used to treat life-threatening racyclines that are more lipophilic than the older tetracyclines.
infections caused by gram-positive bacteria that are resistant to Doxycycline and minocycline also have antiinflammatory and
other antimicrobial drugs, especially methicillin-resistant and immunomodulatory properties that result from inhibition of
vancomycin-resistant staphylococci. Linezolid binds to the 50S inducible nitric oxide synthase and proinflammatory cytokines
ribosome and prevents formation of the initiation complex for such as TNF-α.91 These properties have led to the use of tet-
protein synthesis.89 This is a unique mechanism, because other racyclines for treatment of immune-mediated dermatopathies
protein synthesis inhibitors interfere with polypeptide exten- in dogs92 and plasmacytic pododermatitis in cats.93 Delayed-
sion. Resistance to other protein synthesis inhibitors does not release formulations that produce subantimicrobial doses have
correlate with resistance to linezolid. Rarely, resistance to line- been developed for use in human patients with the inflamma-
zolid is due to modification of the drug’s target site, but this tory skin disease rosacea. Although development of tetracycline
is extremely rare because several step mutations are necessary resistance in bacterial flora has not been documented in associa-
before resistance can occur. The high cost of oral linezolid cur- tion with the use of these preparations, the possibility has not
rently limits its application in small animals. Currently, its use been ruled out.94
has been limited to dogs and cats with resistant staphylococ- Resistance to tetracyclines primarily results from porin
cal or enterococcal infections for which other antibiotics are mutations that exclude tetracyclines from the bacterial cell, and
not effective or have produced adverse effects. Linezolid has increased drug efflux, mediated by the tetK gene. Some bacte-
favorable pharmacokinetics in dogs,90 with nearly 100% oral ria with tetK may still be susceptible to minocycline. Resistance
absorption, which is not influenced by food (Table 8-14). The mediated by tetM confers resistance to all tetracyclines.95 In
pharmacokinetics in cats has not been published. Because line- diagnostic laboratories, tetracycline is used to assess the sus-
zolid is bacteriostatic, plasma concentrations should be main- ceptibility of bacteria to all other tetracyclines except the gly-
tained above the MIC throughout the dosing interval. cylcycline antibiotic tigecycline, a new antibiotic derived from
In human patients, adverse effects include vomiting, diar- minocycline that is reserved for treatment of multidrug-resistant
rhea, and, rarely, thrombocytopenia or reversible pure red bacterial infections.96 In addition, resistance to doxycycline
cell aplasia (PRCA). Linezolid is a type A monoamine oxidase does not always imply resistance to minocycline.97
inhibitor and so can interact with serotonin reuptake inhibi-
tors.92 Linezolid appears to be well tolerated by dogs and cats. Clinical Use
Despite the concern about monoamine oxidase inhibition and Doses are shown in Table 8-15. Doxycycline is the most com-
bone marrow suppression in human patients, these effects have monly used tetracycline in dogs and cats and has fewer adverse
not yet been documented in dogs or cats, although one of the effects when compared with other tetracyclines. It is the drug
authors is aware of a cat that developed reversible PRCA after of choice for treatment of a variety of tick-borne infectious dis-
treatment with linezolid. eases, feline chlamydiosis, salmon poisoning disease, and lep-
tospirosis. The long half-life of doxycycline permits once-daily
Tetracyclines dosing, although twice-daily dosing is frequently recommended.
Mechanism of Action Doxycycline is well absorbed from the gastrointestinal tract and
Tetracyclines are bacteriostatic, time-dependent antibiotics that distributed to a variety of tissues, including the lung, bronchial
have a broad spectrum of activity that includes gram-positive secretions, liver, kidney, and, to some extent, the CNS. If doxy-
and gram-negative bacteria, some anaerobes, and atypical and cycline is not available, minocycline can be used as a substitute.
intracellular pathogens such as spirochetes, Mycoplasma spp., Minocycline is more lipophilic than doxycycline, it is better
and rickettsiae. In contrast to the macrolides and clindamycin, absorbed orally, and can be administered at similar doses as
which bind to the 50S ribosomal subunit, tetracyclines inhibit doxycycline. Absorption of other tetracyclines from the gastroin-
bacterial protein synthesis by binding to the 30S subunit. Thus, testinal tract is not as efficient. All tetracyclines are concentrated
resistance to macrolides does not imply resistance to tetracy- in bile. Sufficient drug is excreted in the urine to permit treatment
clines. Examples of tetracyclines used to treat dogs and cats are of UTIs caused by bacteria that are resistant to other drugs.
CHAPTER 8  Antibacterial Drugs 83

TABLE 8-15
Suggested Tetracycline Dosages for Small Animals
Dose Interval CLSI Breakpoints
Drug (mg/kg) (hours) Species Route Comments (µg/mL)
Tetracycline 15-20 8 D, C PO Do not mix with food containing ≤2 for streptococci, ≤4 for other
4.4-11 IV, IM cations such as calcium, zinc, susceptible bacteria. Consider
magnesium, iron, aluminum. use of ≤1 for susceptible bacteria.
Oxytetracycline 20 12 D, C PO See tetracycline. See doxycycline.
7.5-10 IV
Doxycycline 5 12 D, C PO, IV For IV administration, dilute in ≤2 for streptococci, ≤0.25 for
10 24 100 to 1000 mL of LRS or 5% Staphylococcus pseudinterme-
dextrose and administer over dius, ≤4 for other susceptible
1 to 2 hours. bacteria
Minocycline 5-12.5 12 D, C PO See doxycycline, but the pharma-
cokinetics of minocycline in dogs
and cats require further study.

C, Cats; CLSI, Clinical and Laboratory Standards Institute; D, dogs; LRS, lactated Ringer solution.

Adverse Effects
The most common adverse effects of tetracyclines in dogs and
cats are vomiting, decreased appetite, nausea, and diarrhea.
These may be reduced when doxycycline hyclate tablets are
administered intact (rather than being split or crushed) and by
administering doxycycline with food. Administration of other
tetracyclines with food can lead to significantly reduced drug
absorption, because of extensive binding with divalent and
trivalent cations in the gastrointestinal tract. The hyclate salt
of doxycycline (doxycycline hydrochloride) has been associ-
ated with esophagitis and esophageal strictures in cats (Figure
8-4),98 and life-threatening esophageal ulceration in a dog.99
This is also a complication in people.100 A bolus of water or
food should be administered immediately after dosing in order
to prevent this complication, or a suspension rather than tablets
A should be used. Compounded, high-concentration (e.g., 33.3
mg/mL or 167 mg/mL) suspensions in aqueous vehicles have
limited stability; suspensions in some vehicles do not retain their
potency beyond 7 days.101
In dogs and cats with renal failure, tetracyclines other than
doxycycline should be avoided. Doxycycline is excreted in the
intestinal tract in this situation. Tetracyclines other than dox-
ycycline can interfere with bone growth and cause significant
and permanent gray-brown or yellow discoloration of the teeth
­(Figure 8-5), and so their use is not recommended in puppies
and kittens. Doxycycline is not likely to produce this effect
because it binds calcium less avidly. As a result, the use of dox-
ycycline in human pediatric medicine is now accepted.102 All
tetracyclines, including doxycycline, have the potential to cause
hepatic failure and renal tubular necrosis. Cutaneous reactions
occur rarely in animals treated with tetracycline, but have been
reported in humans.
B Intravenous preparations of doxycycline are available but
FIGURE 8-4  A, Esophageal stricture in a 9-year-old female spayed domestic shorthair can be considerably more expensive than the oral formulations.
that had been treated with doxycycline for possible hemoplasmosis. Regurgitation after They must be given over 1 to 2 hours. Thrombophlebitis can
eating commenced shortly after treatment was initiated. B, The stricture was treated via occur at the site of injection, and anaphylactic shock has been
repeated balloon dilatation; the image is immediately after the ballooning procedure was reported following parenteral administration of tetracycline to
performed. a dog.103
84 SECTION 2  Antiinfective Therapy

18. Svetitsky S, Leibovici L, Paul M. Comparative efficacy and safety


of vancomycin versus teicoplanin: systematic review and meta-
analysis. Antimicrob Agents Chemother. 2009;53:4069-4079.
19. Robicsek A, Jacoby GA, Hooper DC. The worldwide emergence
of plasmid-mediated quinolone resistance. Lancet Infect Dis.
2006;6:629-640.
20. Phillips I, King A. Comparative activity of the 4-quinolones. Rev
Infect Dis. 1988;10(Suppl 1):S70-S76.
21. Rubin J, Walker RD, Blickenstaff K, et  al. Antimicrobial resis-
tance and genetic characterization of fluoroquinolone resistance
of Pseudomonas aeruginosa isolated from canine infections. Vet
Microbiol. 2008;131:164-172.
22. Silley P, Stephan B, Greife HA, et al. Comparative activity of pra-
dofloxacin against anaerobic bacteria isolated from dogs and cats.
J Antimicrob Chemother. 2007;60:999-1003.
23. Papich MG. Ciprofloxacin pharmacokinetics and oral absorption
of generic tablets in dogs. Am J Vet Res. 2012;73:1085-1091.
24. Frazier DL, Thompson L, Trettien A, et al. Comparison of fluoro-
quinolone pharmacokinetic parameters after treatment with mar-
FIGURE 8-5  Yellow discoloration of the teeth in a “puppy mill” puppy that was pre- bofloxacin, enrofloxacin, and difloxacin in dogs. J Vet Pharmacol
sumably treated with tetracycline antibiotics. Ther. 2000;23:293-302.
25. Cole LK, Papich MG, Kwochka KW, et  al. Plasma and ear tis-
sue concentrations of enrofloxacin and its metabolite cipro-
floxacin in dogs with chronic end-stage otitis externa after
REFERENCES
intravenous administration of enrofloxacin. Vet Dermatol. 2009;
1. Silhavy TJ, Kahne D, Walker S. The bacterial cell envelope. Cold 20:51-59.
Spring Harb Perspect Biol. 2010;2:a000414. 26. Mori K, Maru C, Takasuna K. Characterization of histamine
2. Gold HS, Moellering Jr RC. Antimicrobial-drug resistance. release induced by fluoroquinolone antibacterial agents in-vivo
N Engl J Med. 1996;335:1445-1453. and in-vitro. J Pharm Pharmacol. 2000;52:577-584.
3. Bush K, Jacoby GA. Updated functional classification of beta- 27. Christ W. Central nervous system toxicity of quinolones: human
lactamases. Antimicrob Agents Chemother. 2010;54:969-976. and animal findings. J Antimicrob Chemother. 1990;26(Suppl
4. Papich MG. Saunders Handbook of Veterinary Drugs: Small and B):219-225.
Large Animal. 3rd ed. St. Louis, MO: Saunders; 2011. 28. Wiebe V, Hamilton P. Fluoroquinolone-induced retinal degenera-
5. Stapleton PD, Taylor PW. Methicillin resistance in Staphylococcus tion in cats. J Am Vet Med Assoc. 2002;221:1568-1571.
aureus: mechanisms and modulation. Sci Prog. 2002;85:57-72. 29. Ford MM, Dubielzig RR, Giuliano EA, et al. Ocular and systemic
6. Fass RJ, Copelan EA, Brandt JT, et  al. Platelet-mediated manifestations after oral administration of a high dose of enro-
bleeding caused by broad-spectrum penicillins. J Infect Dis. floxacin in cats. Am J Vet Res. 2007;68:190-202.
1987;155:1242-1248. 30. Ramirez CJ, Minch JD, Gay JM, et al. Molecular genetic basis for
7. European Committee on Antimicrobial Susceptibility Testing fluoroquinolone-induced retinal degeneration in cats. Pharmaco-
(EUCAST). MIC Distributions. 2011. http://www.eucast.org/mic_ genet Genomics. 2011;21:66-75.
distributions/. Last accessed May 12, 2012. 31. Messias A, Gekeler F, Wegener A, et al. Retinal safety of a new
8. Williams KJ, Bax RP, Brown H, et  al. Antibiotic treatment fluoroquinolone, pradofloxacin, in cats: assessment with electro-
and associated prolonged prothrombin time. J Clin Pathol. retinography. Doc Ophthalmol. 2008;116:177-191.
1991;44:738-741. 32. Rosanova MT, Lede R, Capurro H, et  al. Assessing fluoroqui-
9. Deldar A, Lewis H, Bloom J, et  al. Cephalosporin-induced nolones as risk factor for musculoskeletal disorders in children:
changes in the ultrastructure of canine bone marrow. Vet Pathol. a systematic review and meta-analysis. Arch Argent Pediatr.
1988;25:211-218. 2010;108:524-531.
10. Asbel LE, Levison ME. Cephalosporins, carbapenems, and mono- 33. Sansone JM, Wilsman NJ, Leiferman EM, et al. The effect of fluo-
bactams. Infect Dis Clin North Am. 2000;14:435-447, ix. roquinolone antibiotics on growing cartilage in the lamb model.
11. Zhanel GG, Wiebe R, Dilay L, et al. Comparative review of the J Pediatr Orthop. 2009;29:189-195.
carbapenems. Drugs. 2007;67:1027-1052. 34. Committee on Infectious Diseases. The use of systemic fluoroqui-
12. Bratu S, Landman D, Alam M, et al. Detection of KPC carbapenem- nolones. Pediatrics. 2006;118:1287-1292.
hydrolyzing enzymes in Enterobacter spp. from Brooklyn, New 35. Narayanan S, Hunerbein A, Getie M, et al. Scavenging properties
York. Antimicrob Agents Chemother. 2005;49:776-778. of metronidazole on free oxygen radicals in a skin lipid model
13. Marra A. NDM-1: a local clone emerges with worldwide aspira- system. J Pharm Pharmacol. 2007;59:1125-1130.
tions. Future Microbiol. 2011;6:137-141. 36. Jergens AE, Crandell J, Morrison JA, et  al. Comparison of
14. Peleg AY, Hooper DC. Hospital-acquired infections due to gram- oral prednisone and prednisone combined with metronida-
negative bacteria. N Engl J Med. 2010;362:1804-1813. zole for induction therapy of canine inflammatory bowel dis-
15. Bailiff NL, Westropp JL, Jang SS, et al. Corynebacterium urealyti- ease: a randomized-controlled trial. J Vet Intern Med. 2010;24:
cum urinary tract infection in dogs and cats: 7 cases (1996-2003). 269-277.
J Am Vet Med Assoc. 2005;226:1676-1680. 37. Lofmark S, Edlund C, Nord CE. Metronidazole is still the drug
16. Briscoe KA, Barrs VR, Lindsay S, et  al. Encrusting cystitis in a of choice for treatment of anaerobic infections. Clin Infect Dis.
cat secondary to Corynebacterium urealyticum infection. J Feline 2010;50(Suppl 1):S16-S23.
Med Surg. 2010;12:972-977. 38. Evans J, Levesque D, Knowles K, et al. Diazepam as a treatment
17. Abbott Y, Kirby BM, Karczmarczyk M, et  al. High-level gen- for metronidazole toxicosis in dogs: a retrospective study of 21
tamicin-resistant and vancomycin-resistant Enterococcus fae- cases. J Vet Intern Med. 2003;17:304-310.
cium isolated from a wound in a dog. J Small Anim Pract. 39. Olson EJ, Morales SC, McVey AS, et al. Putative metronidazole
2009;50:194-197. neurotoxicosis in a cat. Vet Pathol. 2005;42:665-669.
CHAPTER 8  Antibacterial Drugs 85

40. Moosa AN, Perkins D. Neurological picture. MRI of metronida- 61. Cribb AE, Spielberg SP. An in vitro investigation of predisposition
zole induced cerebellar ataxia. J Neurol Neurosurg Psychiatry. to sulphonamide idiosyncratic toxicity in dogs. Vet Res Commun.
2010;81:754-755. 1990;14:241-252.
41. Sekis I, Ramstead K, Rishniw M, et  al. Single-dose pharmaco- 62. Ramirez MS, Tolmasky ME. Aminoglycoside modifying enzymes.
kinetics and genotoxicity of metronidazole in cats. J Feline Med Drug Resist Updat. 2010;13:151-171.
Surg. 2009;11:60-68. 63. Sykes JE, Mapes S, Lindsay LL, et  al. Corynebacterium ulcer-
42. Jenkins DJ, Kendall CW, Hamidi M, et al. Effect of antibiotics as ans bronchopneumonia in a dog. J Vet Intern Med. 2010;24:
cholesterol-lowering agents. Metabolism. 2005;54:103-112. 973-976.
43. Cavusoglu C, Karaca-Derici Y, Bilgic A. In-vitro activity of 64. Gookin JL, Riviere JE, Gilger BC, et  al. Acute renal failure in
rifabutin against rifampicin-resistant Mycobacterium tuberculo- four cats treated with paromomycin. J Am Vet Med Assoc.
sis isolates with known rpoB mutations. Clin Microbiol Infect. 1999;215:1821-1823, 1806.
2004;10:662-665. 65. Lopez-Novoa JM, Quiros Y, Vicente L, et al. New insights into
44. Dutta A, Schwarzwald HL, Edwards MS. Disseminated bar- the mechanism of aminoglycoside nephrotoxicity: an integrative
tonellosis presenting as neuroretinitis in a young adult with point of view. Kidney Int. 2011;79:33-45.
human immunodeficiency virus infection. Pediatr Infect Dis J. 66. Cowan RH, Jukkola AF, Arant Jr BS. Pathophysiologic evidence
2010;29:675-677. of gentamicin nephrotoxicity in neonatal puppies. Pediatr Res.
45. Liston TE, Koehler JE. Granulomatous hepatitis and necrotizing 1980;14:1204-1211.
splenitis due to Bartonella henselae in a patient with cancer: case 67. Rao SC, Ahmed M, Hagan R. One dose per day compared to
report and review of hepatosplenic manifestations of bartonella multiple doses per day of gentamicin for treatment of suspected
infection. Clin Infect Dis. 1996;22:951-957. or proven sepsis in neonates. Cochrane Database Syst Rev.
46. De Vriese AS, Robbrecht DL, Vanholder RC, et  al. Rifampicin- 2006:CD005091.
associated acute renal failure: pathophysiologic, immunologic, 68. Grauer GF, Greco DS, Behrend EN, et al. Effects of dietary protein
and clinical features. Am J Kidney Dis. 1998;31:108-115. conditioning on gentamicin-induced nephrotoxicosis in healthy
47. Reitman ML, Chu X, Cai X, et  al. Rifampin’s acute inhibitory male dogs. Am J Vet Res. 1994;55:90-97.
and chronic inductive drug interactions: experimental and model- 69. Papich MG, Riviere JE. Aminoglycoside antibiotics. In: Riviere JE,
based approaches to drug-drug interaction trial design. Clin Phar- Papich MG, eds. Veterinary Pharmacology and Therapeutics. 9th
macol Ther. 2011;89:234-242. ed. Ames, IA: Wiley-Blackwell; 2009:915-935.
48. Then RL. Mechanisms of resistance to trimethoprim, the sul- 70. Gooi A, Hochman J, Wellman M, et al. Ototoxic effects of single-
fonamides, and trimethoprim-sulfamethoxazole. Rev Infect Dis. dose versus 19-day daily-dose gentamicin. J Otolaryngol Head
1982;4:261-269. Neck Surg. 2008;37:664-667.
49. Chretin JD, Rassnick KM, Shaw NA, et  al. Prophylactic trime- 71. Singh YN, Marshall IG, Harvey AL. Some effects of the amino-
thoprim-sulfadiazine during chemotherapy in dogs with lym- glycoside antibiotic amikacin on neuromuscular and autonomic
phoma and osteosarcoma: a double-blind, placebo-controlled transmission. Br J Anaesth. 1978;50:109-117.
study. J Vet Intern Med. 2007;21:141-148. 72. Forsyth SF, Ilkiw JE, Hildebrand SV. Effect of gentamicin admin-
50. Jacoby GA. Perils of prophylaxis. N Engl J Med. 1982;306:43-44. istration on the neuromuscular blockade induced by atracurium in
51. Weese JS, Blondeau JM, Boothe D, et  al. Antimicrobial use cats. Am J Vet Res. 1990;51:1675-1678.
guidelines for treatment of urinary tract disease in dogs and 73. Martinez EA, Mealey KL, Wooldridge AA, et  al. Pharmacoki-
cats: antimicrobial guidelines working group of the international netics, effects on renal function, and potentiation of atracurium-
society for companion animal infectious diseases. Vet Med Int. induced neuromuscular blockade after administration of a high
2011;2011:263768. dose of gentamicin in isoflurane-anesthetized dogs. Am J Vet Res.
52. Nuttall TJ, Malham T. Successful intravenous human immuno- 1996;57:1623-1626.
globulin treatment of drug-induced Stevens-Johnson syndrome in 74. Harnett MT, Chen W, Smith SM. Calcium-sensing receptor: a
a dog. J Small Anim Pract. 2004;45:357-361. high-affinity presynaptic target for aminoglycoside-induced weak-
53. Noli C, Koeman JP, Willemse T. A retrospective evaluation of ness. Neuropharmacology. 2009;57:502-505.
adverse reactions to trimethoprim-sulphonamide combinations in 75. Papich MG. Therapeutic drug monitoring. In: Riviere JE, Papich
dogs and cats. Vet Q. 1995;17:123-128. MG, eds. Veterinary pharmacology and therapeutics. 9th ed.
54. Weiss DJ, Adams LG. Aplastic anemia associated with trime- Ames, IA: Wiley-Blackwell; 2009:1328.
thoprim-sulfadiazine and fenbendazole administration in a dog. 76. De Renzo A, Formisano S, Rotoli B. Bone marrow aplasia and
J Am Vet Med Assoc. 1987;191:1119-1120. thiamphenicol. Haematologica. 1981;66:98-104.
55. Trepanier LA, Danhof R, Toll J, et al. Clinical findings in 40 dogs 77. Schwarz S, Kehrenberg C, Doublet B, et  al. Molecular basis of
with hypersensitivity associated with administration of potenti- bacterial resistance to chloramphenicol and florfenicol. FEMS
ated sulfonamides. J Vet Intern Med. 2003;17:647-652. Microbiol Rev. 2004;28:519-542.
56. Trepanier LA. Idiosyncratic toxicity associated with potenti- 78. Wiholm BE, Kelly JP, Kaufman D, et al. Relation of aplastic anae-
ated sulfonamides in the dog. J Vet Pharmacol Ther. 2004;27: mia to use of chloramphenicol eye drops in two international case-
129-138. control studies. BMJ. 1998;316:666.
57. Vasilopulos RJ, Mackin A, Lavergne SN, et  al. Nephrotic 79. Rayner SA, Buckley RJ. Ocular chloramphenicol and aplastic
syndrome associated with administration of sulfadimethox- anaemia. Is there a link? Drug Saf. 1996;14:273-276.
ine/ormetoprim in a dobermann. J Small Anim Pract. 2005;46: 80. Giamarellos-Bourboulis EJ. Macrolides beyond the conventional
232-236. antimicrobials: a class of potent immunomodulators. Int J Antimi-
58. Brenner K, Harkin K, Schermerhorn T. Iatrogenic, sulfonamide- crob Agents. 2008;31:12-20.
induced hypothyroid crisis in a Labrador Retriever. Aust Vet J. 81. Schultz MJ. Macrolide activities beyond their antimicrobial
2009;87:503-505. effects: macrolides in diffuse panbronchiolitis and cystic fibrosis.
59. Twedt DC, Diehl KJ, Lappin MR, et  al. Association of hepatic J Antimicrob Chemother. 2004;54:21-28.
necrosis with trimethoprim sulfonamide administration in 4 dogs. 82. Sivapalasingham S, Steigbigel N. Macrolides, clindamycin and
J Vet Intern Med. 1997;11:20-23. ketolides. In: Mandell GL, Bennett JE, Dolin R, eds. Mandell,
60. Osborne CA, Lulich JP, Bartges JW, et  al. Drug-induced uroli- Douglas, and Bennett’s Principles and Practice of Infectious Dis-
thiasis. Vet Clin North Am Small Anim Pract. 1999;29:251-266, eases. 7th ed. Philadelphia, PA: Churchill Livingstone Elsevier;
xiv. 2011:427-448.
86 SECTION 2  Antiinfective Therapy

83. Piscitelli SC, Danziger LH, Rodvold KA. Clarithromycin 93. Bettenay SV, Mueller RS, Dow K, et al. Prospective study of the
and azithromycin: new macrolide antibiotics. Clin Pharm. treatment of feline plasmacytic pododermatitis with doxycycline.
1992;11:137-152. Vet Rec. 2003;152:564-566.
84. Westermarck E, Skrzypczak T, Harmoinen J, et  al. Tylosin- 94. McKeage K, Deeks ED. Doxycycline 40 mg capsules (30 mg
responsive chronic diarrhea in dogs. J Vet Intern Med. 2005; immediate-release/10 mg delayed-release beads): anti-inflammatory
19:177-186. dose in rosacea. Am J Clin Dermatol. 2010;11:217-222.
85. Ruch-Gallie RA, Veir JK, Spindel ME, et al. Efficacy of amoxicil- 95. Thaker M, Spanogiannopoulos P, Wright GD. The tetracycline
lin and azithromycin for the empirical treatment of shelter cats resistome. Cell Mol Life Sci. 2010;67:419-431.
with suspected bacterial upper respiratory infections. J Feline Med 96. Pankey GA. Tigecycline. J Antimicrob Chemother. 2005;
Surg. 2008;10:542-550. 56:470-480.
86. Peeters TL. Erythromycin and other macrolides as prokinetic 97. Weese JS, Sweetman K, Edson H, et al. Evaluation of minocycline
agents. Gastroenterology. 1993;105:1886-1899. susceptibility of methicillin-resistant Staphylococcus pseudinter-
87. Beatty JA, Swift N, Foster DJ, et al. Suspected clindamycin-asso- medius. Vet Microbiol. 2013;162:968-971.
ciated oesophageal injury in cats: five cases. J Feline Med Surg. 98. German AJ, Cannon MJ, Dye C, et  al. Oesophageal strictures
2006;8:412-419. in cats associated with doxycycline therapy. J Feline Med Surg.
88. Glanemann B, Hildebrandt N, Schneider MA, et  al. Recurrent 2005;7:33-41.
single oesophageal stricture treated with a self-expanding stent in 99. Sykes JE, Bailiff NL, Ball LM, et al. Identification of a novel hemo-
a cat. J Feline Med Surg. 2008;10:505-509. tropic mycoplasma in a splenectomized dog with hemic neoplasia.
89. Livermore DM. Linezolid in  vitro: mechanism and antibacterial J Am Vet Med Assoc. 2004;224:1945-1951, 1930-1931.
spectrum. J Antimicrob Chemother. 2003;51(Suppl 2):ii9-i16. 100. Morris TJ, Davis TP. Doxycycline-induced esophageal ulceration
90. Beekmann SE, Gilbert DN, Polgreen PM. Toxicity of extended in the U.S. Military service. Mil Med. 2000;165:316-319.
courses of linezolid: results of an Infectious Diseases Society of 101. Papich MG, Davidson G. Doxycycline potency after storage in a
America Emerging Infections Network survey. Diagn Microbiol compounded formulation for animals. American College of Vet-
Infect Dis. 2008;62:407-410. erinary Internal Medicine Forum. 2011. Denver, CO, USA.
91. Leite LM, Carvalho AG, Ferreira PL, et  al. Anti-inflammatory 102. Volovitz B, Shkap R, Amir J, et  al. Absence of tooth staining
properties of doxycycline and minocycline in experimental mod- with doxycycline treatment in young children. Clin Pediatr (Phila).
els: an in vivo and in vitro comparative study. Inflammopharma- 2007;46:121-126.
cology. 2011;19:99-110. 103. Ward GS, Guiry CC, Alexander LL. Tetracycline-induced anaphy-
92. White SD, Rosychuk RA, Reinke SI, et al. Use of tetracycline and lactic shock in a dog. J Am Vet Med Assoc. 1982;180:770-771.
niacinamide for treatment of autoimmune skin disease in 31 dogs.
J Am Vet Med Assoc. 1992;200:1497-1500.
CHAPTER 9

Antifungal Drugs
Jane E. Sykes and Mark G. Papich

w KEY POINTS
• C  ompared to antibacterial drugs, the range of antifungal drug done because of variable availability of clinical assays and
classes available is very limited, with most systemic treatment uncertainty in interpretation of the measured concentrations.
accomplished with the azole group of medications. The azoles • Fungal infections often require prolonged treatment durations.
have a similar mechanism of action and share other properties This may be costly for some pet owners and can increase the
such as spectrum of activity, pharmacokinetics, tissue penetra- risk of adverse effects to pets.
tion, and adverse effect profiles. Proper understanding of these • Fungal organisms that are resistant to certain antifungal drugs
factors is most likely to result in successful treatment with mini- are increasingly recognized. Although antifungal suscepti-
mal adverse effects. bility testing can identify resistant organisms, this is often
• Drug interactions occur with many antifungal drugs. not performed because there are no standards for veterinary
• Although therapeutic drug monitoring would improve treat- pathogens and drugs used in veterinary medicine, and few
ment with the azole antifungals and 5-flucytosine, this is rarely laboratories perform these tests.

INTRODUCTION with antifungal drugs must often be continued for months, and
in some animals it may be weeks before clinical improvement is
The use of antifungal drugs in human and veterinary medicine evident. Sometimes, disease worsens in the first week of treat-
has increased in recent years. In humans, increased suscepti- ment, because of the host’s inflammatory response to killed
bility to fungal infections has arisen because of immunosup- organisms. This may have severe consequences for the host
pression secondary to HIV infection or treatment with potent when there is extensive involvement of the pulmonary paren-
immunosuppressive drugs. In animals, opportunistic fungal chyma or the central nervous system (CNS). Concurrent treat-
infections also result from immunosuppression, and the impor- ment with a nonsteroidal antiinflammatory drug (NSAIDs) or,
tance of fungal infections in dogs and cats is increasingly recog- when CNS involvement is present, judicious use of a short course
nized. Antifungal drugs used in dogs and cats are generally not of antiinflammatory glucocorticoids may improve outcome in
approved for use in these species. Therefore, veterinarians use these situations.1
human antifungal drugs off-label, with indications and dosing The prognosis for animals with disseminated mold infec-
protocols often extrapolated from the human use. Because of tions that have reversible underlying immunosuppression (such
differences in pharmacokinetics among species for these drugs, as drug-induced immunosuppression) may be better than the
and differences in susceptibility to adverse effects in dogs and prognosis for animals with no apparent underlying immuno-
cats compared to people, this approach is not optimal. Fortu- suppressive disease. This is because the latter group of animals
nately, the clinical experience and pharmacokinetic data have are likely to have underlying (irreversible) genetic defects in
expanded for these human drugs, which will improve anti- immunity.
fungal treatment in animals. Efforts also have focused on the
development of new, less toxic, and more efficacious antifungal Azole Antifungals
drugs. This chapter reviews the classification, mechanisms of
action, spectrum of activity, resistance mechanisms, tissue pen- Mechanism of Action, Classification,
etration, clinical use, and adverse effects of the major systemic and Spectrum of Activity
antifungal drugs used to treat dogs and cats. Topical antifungal Azole antifungal drugs inhibit sterol 14α-demethylase, a cyto-
treatments are reviewed in the relevant chapters of this book. chrome P450–dependent fungal enzyme involved in synthesis of
Basic principles of antimicrobial drug treatment are discussed ergosterol, a key component of the fungal cell wall, from lanos-
in Chapter 6. terol. The result is the accumulation of 14α-methylsterols, which
Antifungal drug treatment is often more prolonged than disrupt the fungal cell membrane (Figure 9-1). The majority of
antibacterial drug treatment. One reason for this difference is the adverse effects and drug interactions observed with these
that fungal organisms grow more slowly, and the drugs used drugs relate to the cross-inhibition of mammalian P450 enzymes.
(primarily azoles) are fungistatic, not fungicidal. Therefore, long- All of the azole antifungals have the potential to be teratogenic,
term treatment is needed to inhibit fungal growth and allow for and their use should be avoided in pregnancy.
the animal’s immune system, which is often compromised, to Azole antifungals are classified as imidazoles or triazoles
eradicate the infection. Treatment of animals with deep mycoses based on whether they possess two or three nitrogen molecules

87
88 SECTION 2  Antiinfective Therapy

FIGURE 9-1  Mechanism of action of antifungal drugs. 5FC, 5-flucytosine; 5FUTP, 5-fluorouridine triphosphate; 5FdUMP, 5-fluorodeoxyuridine monophosphate; A, amphotericin B;
E, ergosterol. Modified from Rex JH, Stevens DA. Systemic antifungal agents. In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, Douglas and Bennett’s Principles and Practice of Infectious
Diseases, 7 ed. Philadelphia, PA: Churchill Livingstone Elsevier; 2011:549-563.

in their azole ring. Ketoconazole, enilconazole, and clotrimazole Increasingly, it is apparent that resistance to one azole does not
are imidazoles. The latter two drugs have poor oral bioavailabil- always imply resistance to other azole antifungal drugs.
ity and are used topically in veterinary medicine for the treat-
ment of superficial mycoses (see Chapters 65 and 67). Triazole Ketoconazole
antifungal drugs, such as itraconazole and fluconazole, are more Spectrum of Activity and Clinical Use
slowly metabolized and have less impact on mammalian sterol The use of ketoconazole has largely been replaced by itracon-
synthesis than do imidazoles. Imidazoles and triazoles have azole for treatment of many mycoses, because of the greater
been widely used to treat a variety of mycoses, which include toxicity and reduced efficacy of ketoconazole when compared
candidiasis, cryptococcosis, blastomycosis, histoplasmosis, coc- with triazole antifungal drugs. Because of its low cost, keto-
cidioidomycosis, dermatophytosis, sporotrichosis, and aspergil- conazole continues to be used in veterinary medicine when the
losis (Tables 9-1 and 9-2). cost of other antifungal drugs is prohibitive to the client, and it
Unfortunately, resistance to azole antifungal drugs has remains efficacious for treatment of Malassezia dermatitis4 and
emerged among some fungi. Resistance results from mutations feline nasal and cutaneous cryptococcosis. The absorption of
in the gene encoding the demethylase enzyme, increased produc- ketoconazole is improved when it is administered with food,
tion of C-14α demethylase, and increased azole efflux by fungal but it is inhibited by concurrent use of antacids. Ketoconazole
cell membrane transporters. Methods for in  vitro susceptibil- is highly protein bound and is metabolized extensively by the
ity testing have become more standardized,2,3 with improved liver. Nevertheless, moderate hepatic dysfunction does not alter
correlation between the results of in  vitro susceptibility test- blood levels of ketoconazole. Inactive products are excreted in
ing and clinical response, but breakpoints (criteria that define bile and, to a lesser extent, the urine. Because of poor CNS pen-
susceptible versus resistant minimum inhibitory concentration etration, it is ineffective for treatment of meningeal cryptococ-
values) are still needed for many drug-fungus combinations. cosis and aspergillosis.5
CHAPTER 9  Antifungal Drugs 89

TABLE 9-1
Spectrum of Activity and Tissue Distribution of Different Antifungal Drugs
Drug Spectrum of Activity Tissue Distribution
Ketoconazole Dimorphic fungi, Malassezia. Ineffective for aspergillosis Skin, bone, joint, lung. Poor CNS penetration
Itraconazole Dimorphic fungi and molds, Malassezia spp. Skin, bone, lung. May enter the CNS and
eye with inflammation
Fluconazole Some Candida spp., Malassezia spp., some dimorphic fungi. Widely distributed, including to the skin,
Poor activity against molds. Aspergillus spp. are intrinsi- lung, CNS, urine, and eye
cally resistant
Voriconazole Dimorphic fungi, yeasts, and molds with the exception of CNS, eye, lung, bone
Sporothrix schenckii and zygomycetes
Posaconazole Dimorphic fungi, yeasts, and molds including zygomycetes Widely distributed
Amphotericin B Broad spectrum. Also active against Leishmania Limited penetration of the CNS and eye
5-Flucytosine Cryptococcus and Candida spp. Widely distributed, which includes the CNS,
eye, and urine
Griseofulvin Dermatophytes Concentrates in skin
Terbinafine Activity highest for dermatophytes. To a lesser extent may Concentrates in skin and hair
have activity against other dimorphic and filamentous fungi
Caspofungin Candida and Aspergillus spp. Not active against Cryptococcus Widely distributed. Poor penetration of the
spp. or when given alone to treat Coccidioides CNS and eye

CNS, Central nervous system.

TABLE 9-2
Suggested Azole Antifungal Drug Doses for Dogs and Cats
Drug Dose Interval Species Route Comments
(mg/kg) (hours)
Ketoconazole 10 to 15 12 D PO Do not administer to pregnant animals. Monitor liver enzymes monthly
5 to 10 C during treatment. Drug interactions may occur (see text). Inhibits
adrenal function. Antacids impair absorption.
Itraconazole 5 12 to 24 D, C PO Do not administer to pregnant animals. Monitor liver enzymes monthly
during treatment. Drug interactions may occur. Use of the oral suspen-
sion warrants dose reduction to 3 mg/kg. Monitor serum drug concen-
trations after 2 weeks if there is inadequate response to treatment (see
text). Compounded formulations are unstable.
Fluconazole 5 to 10 12 D PO Do not administer to pregnant animals. Monitor liver enzymes monthly
50 mg/cat 12 to 24 C during treatment. Drug interactions may occur.
Voriconazole 4 to 5 12 D PO Do not use in cats. Use cautiously in animals with liver disease. Also see
fluconazole. Consider therapeutic drug monitoring (see text).
Posaconazole 5 to 10 12 to 24 D PO Absorption may be improved when daily dose is split into 2 to 4 doses.
5 24 C Consider therapeutic drug monitoring. Also see fluconazole. Antacids
impair absorption.

C, Cats; D, dogs.

Adverse Effects vomiting, lethargy, increasing activities of serum ALT and


The most common adverse effects of ketoconazole in dogs and ALP, and hyperbilirubinemia. The drug should be discontin-
cats are vomiting, anorexia, lethargy, and diarrhea.6,7 Admin- ued if this occurs and serum chemistry values checked 1 to 2
istration of ketoconazole with food may reduce gastrointes- days later. Hepatitis can occur at any time, and the onset may
tinal adverse effects. Mild increases in the activity of serum be extremely rapid. Pruritis and cutaneous erythema have been
transaminases occur commonly during treatment, but do not reported in fewer than 1% of dogs treated with ketoconazole.6
warrant discontinuation of the drug. Less commonly, ketocon- Lightening of the hair coat color and cataract formation occur
azole causes hepatitis, which may be accompanied by anorexia, rarely.
90 SECTION 2  Antiinfective Therapy

Ketoconazole is a potent inhibitor of mammalian cytochrome


P450 enzymes and efflux transporter proteins such as P-glyco-
protein. It also inhibits testosterone and cortisol synthesis. As a
result, it has been used to treat pituitary-dependent hyperadre-
nocorticism in dogs8 and also to deliberately inhibit the metabo-
lism of cyclosporin, allowing a reduction in dose and, therefore,
cost,9 although there are concerns about whether use of keto-
conazole in this way could contribute antifungal drug resistance
and it increases the risk of adverse drug reactions. Transient
infertility can occur during treatment of intact male animals.
Ketoconazole can interfere with P-glycoprotein transport of
ivermectin, which predisposes dogs to ivermectin toxicity.6,10

Itraconazole
Spectrum of Activity and Clinical Use
Itraconazole is one of the most widely used azoles in veteri-
nary medicine. It has been used to treat blastomycosis, sporo-
trichosis, aspergillosis, coccidioidomycosis, dermatophytosis,
histoplasmosis, phaeohyphomycosis, paecilomycosis, crypto- FIGURE 9-2  Submandibular ulcerative skin lesions in a golden retriever that had
coccosis, and Malassezia infections. It is available in capsules been treated with itraconazole for systemic blastomycosis.
(which contain itraconazole granules) and an oral solution. The
IV solution has been withdrawn.11 Itraconazole in capsules is
best absorbed when given with food because the acid secretion
stimulated with feeding increases the drug solubility, which is Adverse Effects
necessary for dissolution and absorption. However, the oral The most common adverse effects of itraconazole in dogs and
solution is complexed with cyclodextrin to improve solubility, cats are vomiting and anorexia. Division of the total dose for
and administration of food does not influence absorption of twice-daily administration has been associated with decreased
this formulation. Absorption of the oral solution is consistently gastrointestinal signs and improved absorption in human
better than absorption of the capsule formulation, regardless patients.5 Gastrointestinal adverse effects may be more common
of feeding.12 As with ketoconazole, concurrent administration with the oral solution. Mild to moderate increases in serum ALT
of gastric acid suppressants (H2-blockers, proton pump inhibi- activity commonly occur during treatment. Provided these are
tors) may reduce absorption of the oral capsule. In cats, the oral not accompanied by inappetence, treatment need not be discon-
solution is very well absorbed, and dose reduction is indicated tinued. Significant hepatotoxicity, accompanied by anorexia,
to reduce the chance of toxicity (see Table 9-2). In addition to vomiting, and hyperbilirubinemia, is less likely to occur than
hydroxypropyl-β-cyclodextrin, this formulation contains sor- with ketoconazole but has been reported in humans as well as
bitol, propylene glycol, hydrochloric acid, cherry flavor, and cats and dogs.13,17-19 Hepatotoxicity more commonly occurs in
saccharin, which may be unpalatable for cats. In Europe and dogs treated with 10 mg/kg/day.13 Ulcerative skin lesions can
Canada an oral solution for cats is available (Itrafungol, 10 mg/ occur in dogs, especially when doses of 10 mg/kg/day or higher
mL). Compounded formulations prepared by pharmacists have are used (Figure 9-2). Development of hepatitis or severe cuta-
highly variable, poor, and even negligible oral absorption in ani- neous ulceration should prompt discontinuation of the drug. It
mals, in addition to being unstable formulations. As a result, may be possible to reinstitute treatment at a lower dose once the
compounded formulations should never be used. adverse effects have resolved. Occasionally mild, focal cutane-
Like ketoconazole, itraconazole undergoes hepatic metabo- ous ulcerative lesions can resolve spontaneously, without dis-
lism and inhibits metabolism of other P450-dependent drugs continuation of treatment.
(e.g., cisapride, diazepam, cyclosporin). It is the only triazole Itraconazole does not suppress adrenal and testicular func-
antifungal drug that is converted to an active metabolite, tion like ketoconazole, but can inhibit the metabolism of
hydroxylitraconazole.11 Itraconazole is highly (99%) plasma other P450 enzyme-dependent drugs, including cyclosporine,
protein bound and does not appear in urine, cerebrospinal fluid digoxin, cisapride, and vinca alkaloids. It can also interfere
(CSF), or ocular tissues, although penetration of the CSF and with ­P-glycoprotein transport of ivermectin, which results in
eye can occur in the presence of inflammation. Itraconazole increased plasma ivermectin concentrations.20
accumulates in the skin and claws and is the drug of choice
for treatment of dermatophytosis in cats. Itraconazole is also Fluconazole
a good choice for treatment of fungal osteomyelitis. Although Spectrum of Activity and Clinical Use
loading doses have been recommended, these initial high doses Fluconazole is the least active azole antifungal drug and has the
can increase the risk of toxicity and do not appear to improve narrowest spectrum. The activity of fluconazole is limited to
outcome.13 Advanced liver disease increases itraconazole con- some Candida spp., Cryptococcus spp., Malassezia spp., and
centrations. Because of the variable absorption of oral itracon- some dimorphic fungi. Fluconazole has poor activity against
azole, monitoring of steady-state plasma concentrations (e.g., molds, and Aspergillus species are intrinsically resistant to flu-
3 weeks after initiating treatment) may be helpful if clinical conazole. Some Cryptococcus isolates are resistant to flucon-
responses are suboptimal.14 Trough plasma itraconazole con- azole, and resistance can develop during treatment. Itraconazole
centrations should be 0.5 to 1 µg/mL as determined using high- is preferable to fluconazole for treatment of histoplasmosis,
performance liquid chromatography.15,16 blastomycosis, sporotrichosis, and dermatophytosis, although
CHAPTER 9  Antifungal Drugs 91

fluconazole has been successfully used to treat coccidioidomy- is the most potent inhibitor of P450 enzymes. Voriconazole can
cosis, blastomycosis, and Malassezia infections in dogs.19 induce its own metabolism over time.26 Because of its high cost,
Fluconazole is available as tablets, an oral suspension, and adverse effects have not been well documented in dogs. The
as an IV solution. The availability of generic fluconazole has author has observed inappetence, increased serum liver enzyme
greatly reduced its cost and increased its use in veterinary medi- activities, and, uncommonly, CNS signs, including ataxia and
cine. Approved generic fluconazole tablets are bioequivalent to staring. Tachypnea and marked pyrexia after IV infusion was
the brand name formulation in people. Similar bioequivalence is observed in one dog. Cats are extremely sensitive to adverse
likely in animals. Fluconazole is more water-soluble and stable effects of voriconazole and predictably develop inappetence, as
than itraconazole, and the compounded oral suspensions can well as ocular and CNS signs that have included ataxia, pelvic
be used in animals with a 14-day beyond-use-day (BUD). Some limb paresis, mydriasis, apparent blindness, decreased pupillary
compounded preparations may have reduced activity. Flucon- light responses, and a decreased menace response.27 Cardiac
azole is almost completely absorbed after oral administration, arrhythmias and hypokalemia also occur in cats. A safe dose
and bioavailability is not altered by food or gastric acidity. In has not been established for cats.
contrast to itraconazole and ketoconazole, fluconazole is only
weakly protein bound. Because it is more water soluble than Posaconazole
other azoles, fluconazole diffuses into body fluids such as saliva, Posaconazole is an itraconazole analogue that has demonstrated
urine, synovial fluid, and CSF. Therefore, it is the drug of choice good efficacy for treatment of several refractory deep mycoses
for treatment of susceptible meningeal and urinary tract fungal in animals, which includes aspergillosis and mucormycosis in
infections. Renal excretion accounts for more than 90% of the cats and invasive aspergillosis in dogs.25,28-30 It has the broadest
elimination of fluconazole. The dosage should be decreased in spectrum of activity of all the azoles. Its spectrum of activity is
animals with renal failure; this is especially important in patients similar to that of voriconazole, but also includes the zygomyce-
receiving other P450-dependent drugs. tes.11 Posaconazole is available as an oral suspension. As with
itraconazole, its absorption is promoted by the concurrent pres-
Adverse Effects ence of fatty food. Gastric acid suppression can reduce the bio-
Adverse effects of fluconazole are uncommon to rare. When availability of posaconazole.31 Absorption is improved when the
they occur, they are similar to those of itraconazole and include total daily dose is divided into two to four doses.32 Therapeutic
vomiting, anorexia, diarrhea, and increased liver enzymes. Dogs drug monitoring is recommended in human patients; target peak
that fail to tolerate itraconazole may tolerate fluconazole with- concentrations are greater than 1.48 mg/L.15,16 Posaconazole is
out increases in liver enzymes.19 Cutaneous rash, cytopenias, highly protein bound (>95%) and undergoes hepatic metabo-
and retinal edema with impaired vision have been reported lism, with some inhibition of P450 enzymes. Most administered
in human patients.21,22 In contrast to itraconazole, cutaneous posaconazole is eliminated in the feces. Data on CSF concentra-
ulceration has not been reported in dogs. tions of posaconazole are not yet available but it appears to be
effective for treatment of CSF cryptococcosis. Adverse effects
Voriconazole are less common in cats when compared with voriconazole, but
Spectrum of Activity and Clinical Use information is limited at the time of writing.28-30,33
Voriconazole is a second-generation triazole that is derived
from fluconazole. In humans, it is the drug of choice for treat- Other Azole Antifungals
ment of invasive aspergillosis.23 It is also used to treat serious Other azole drugs that are undergoing clinical trials in humans
and refractory mold infections caused by Scedosporium spp., are ravuconazole and isavuconazole. Ravuconazole is a flucon-
Paecilomyces spp., and Fusarium spp. Voriconazole is at least azole derivative with good oral bioavailability. Isavuconazole
as active as itraconazole against veterinary isolates of Crypto- is a water-soluble triazole with a spectrum similar to that of
coccus spp., Candida spp., and Aspergillus fumigatus.24 It is not posaconazole but is available in oral and IV formulations. It is
active against Sporothrix spp. or the zygomycetes. metabolized by P450 enzymes and does not achieve therapeutic
Voriconazole is available as a tablet, an oral suspension, and concentrations in the urine. The CNS penetration of isavucon-
an IV solution in a sulfobutylether–β-cyclodextrin base. Like azole has not yet been evaluated.34
fluconazole, it has excellent oral bioavailability, but its absorp-
tion is significantly reduced in the presence of food. Voricon- Amphotericin B
azole is poorly water soluble and moderately protein bound. It
is extensively metabolized by hepatic cytochrome P450 enzymes Mechanism of Action and Spectrum of Activity
and eliminated into bile. The degree to which active drug enters Amphotericin B (AMB) is a polyene macrolide antibiotic pro-
the urinary tract in dogs is not clear. It has good CNS pen- duced by Streptomyces nodosus. It is closely related to nystatin.
etration. Voriconazole has been used with some success to treat AMB irreversibly binds sterols in fungal cell membranes, form-
systemic mold infections in dogs.25 If therapeutic drug monitor- ing pores or channels with subsequent leakage of ions. Although
ing can be performed, the recommended trough concentration generally considered to be fungistatic, at high doses it may be
target is between 1 and 5 mg/L, and peak concentrations below fungicidal. AMB also possesses immunomodulatory effects; it
6 mg/L.15,16,23 activates macrophages and enhances macrophage-killing capac-
ity. This may help explain its efficacy against pathogens such as
Adverse Effects Pythium insidiosum, which lack cell wall ergosterol.
In humans, adverse effects of voriconazole include reversible Amphotericin B has antifungal activity against all of the
visual effects such as photophobia and blurred vision, hallucina- important small animal fungal pathogens. It is also active
tions, peripheral neuropathies, and photosensitization, as well against Leishmania spp.35,36 In human medicine, AMB is the
as the same toxicities as other triazoles. Of all the triazoles, it preferred drug for treatment of rapidly progressive mycoses,
92 SECTION 2  Antiinfective Therapy

BOX 9-1
Suggested Amphotericin B Infusion Protocols for Dogs and Cats

Formulation Protocol
All formulations Obtain baseline CBC, kidney panel, and UA and ensure adequate hydration before starting treatment.
Recheck a kidney panel before each infusion. Administer three times weekly for 4 weeks or until azotemia is
detected. For some patients, monthly administration of a single dose may be required to maintain remission.
Amphotericin B Reconstitute vial contents in sterile water to 5 mg/mL. Administer 0.9% NaCl IV at 1.5-2 times the cal-
deoxycholate (IV culated maintenance rate for 1 hour before and after AMB treatment. Ensure lines are flushed with D5W
route) before infusing AMB.
Dogs: Transfer 0.5 mg/kg to a 250-mL to 1000-mL bag of D5W (total fluid volume administered should
be based on body weight and ability to tolerate a fluid load). Administer IV over 4-6 hours.
Cats: Dilute 0.25 mg/kg in 30 mL of D5W and administer IV over 30 minutes to 1 hour.
Amphotericin B Transfer 0.5 mg/kg (cats) or 0.8 mg/kg (dogs) to bag containing 400 mL (cats) or 500 mL (dogs) of
deoxycholate (SC 0.45% sodium chloride in 2.5% dextrose. Administer subcutaneously. Fractious cats may need to be
route) sedated or placed in a restraint bag. Sterile injection site abscess formation may occur with this protocol,
especially with concentrations of amphotericin B > 20 mg/L.
Amphotericin Dilute to 1 mg/mL in D5W. Administer calculated dose IV over 1 to 2 hours.
B lipid complex Dogs: 3 mg/kg
(Abelcet) Cats: 1 mg/kg

AMB, Amphotericin B; D5W, 5% dextrose in water; UA, urinalysis.

immunosuppressed hosts, or fungal meningitis. Resistance to potassium supplementation to counteract these may be required.
AMB is rare, although high minimum inhibitory concentrations Loading with sodium before the infusion may decrease nephro-
have been observed for some molds, including some isolates of toxicity.40 Slow administration in a large volume of fluid also
Aspergillus terreus, Paecilomyces spp., and Scedosporium spp., decreases nephrotoxicity. Concurrent use of other nephrotoxic
as well as Sporothrix schenckii. The mechanism of resistance drugs, such as aminoglycosides, should be avoided. In humans,
is not well understood, but it may relate to decreased cell wall AMB-D can also cause fever, chills, headache, nausea, vomit-
ergosterol content.37 ing, and, rarely, cytopenias and anaphylaxis.40 Fever, inappe-
tence, and vomiting also appear to occur in some dogs treated
Clinical Use with AMB-D. Phlebitis can occur at the IV infusion site. Other
Amphotericin B has poor aqueous solubility and is not absorbed adverse effects noted in humans include infusion-related hypo-
from the gastrointestinal tract, so it is formulated for IV infusion tension and hypochromic, normocytic anemia. NSAIDs can be
in lyophilized form as a complex with the bile salt deoxycholate administered to decrease pyrexia during treatment.
(Fungizone, AMB-D) (Box 9-1). The complex forms a colloid in
water. Addition of electrolytes to the solution causes it to aggre- Lipid Formulations of Amphotericin B
gate, so it is administered in 5% dextrose (D5W). It is not neces- Three lipid formulations of AMB are marketed in the United
sary to protect the infusion from light, as recommended in the States. These are less nephrotoxic than AMB-D but are consid-
past. In the bloodstream, AMB dissociates from deoxycholate and erably more expensive. Decreased nephrotoxicity results from a
binds extensively to plasma proteins and cholesterol in membranes reduced rate of transfer of AMB to mammalian cell membranes
throughout tissues. Penetration of the vitreous humor and CSF is and increased drug clearance from the blood by the mononu-
poor, but some animals with CNS infections still respond to treat- clear phagocyte system. This allows administration of a higher
ment. In human patients, AMB-D has been given intrathecally to dose of the drug, sometimes with improved treatment efficacy.
treat fungal meningitis and intraocularly following vitrectomy.38 Nevertheless, renal function should still be monitored.
A protocol for subcutaneous administration of AMB-D has Amphotericin B colloidal dispersion (ABCD, Amphotec)
been reported for treatment of cryptococcosis,39 which may be contains AMB and cholesterol sulfate, which form disc-shaped
less costly than IV administration. Sterile injection site abscesses particles. It is more likely to cause chills, fever, and hypotension
occur in some animals treated using this protocol, so IV admin- in humans than AMB-D (80% compared with 12% for AMB-
istration is preferred if possible. D), so it is given over 3 to 4 hours and with premedication.
Liposomal amphotericin B (Ambisome) is a widely used for-
Adverse Effects mulation for treatment of human patients. It consists of AMB
The major adverse effect of AMB-D is nephrotoxicity, which is and a lipid mixture (phosphatidylcholine, cholesterol, and dis-
dose dependent and transient if it is detected early and the drug tearoyl phosphatidylglycerol). After reconstitution in D5W, it
is discontinued. Both an acute effect caused by renal vasocon- forms 80-nm vesicles. Nephrotoxicity and infusion-related reac-
striction and a chronic cumulative effect from repeated admin- tions are much less common with liposomal amphotericin B than
istration occurs. Renal tubular acidosis, nephrogenic diabetes with ABCD or AMB-D, and the drug appears to reach higher
insipidus, hypokalemia, and hypomagnesemia uncommonly concentrations in the eye.41 In human patients, it is used as sal-
occur with treatment. Rarely, aggressive IV fluid therapy and vage therapy for aspergillosis, cryptococcosis, and candidiasis.
CHAPTER 9  Antifungal Drugs 93

TABLE 9-3
Suggested 5-Flucytosine Drug Doses for Cats
Drug Dose (mg/kg) Interval (hours) Species Route Comments
5-Flucytosine 25 to 50 6 to 8 C PO Monitor CBC. Use cautiously in animals with impaired
renal function. Avoid use in dogs. Serum concentra-
tion monitoring is recommended in humans.

C, Cats.

Amphotericin B lipid complex (ABLC, Abelcet) is a mix- Adverse Effects


ture of AMB, dimyristoyl phosphatidylcholine, and dimyris- Administration of flucytosine should be avoided in dogs, which
toyl phosphatidylglycerol that forms ribbon-like sheets. ABLC often develop a severe (but reversible) drug eruption within 2
is the most common formulation used in veterinary medicine. to 3 weeks of starting treatment. The most common adverse
In humans, nephrotoxicity and infusion-related reactions are effects of flucytosine in cats are myelosuppression and gastroin-
intermediate between AMB-D and liposomal amphotericin B. testinal signs. The CBC should be monitored during treatment,
ABLC has been used in small animals to treat refractory cryp- and therapeutic drug monitoring should be considered. Myelo-
tococcosis and cryptococcal meningitis and disseminated coc- suppression is more likely to occur in human patients receiving
cidioidomycosis, aspergillosis, blastomycosis, histoplasmosis, other myelosuppressive drugs. Other adverse effects in humans
and pythiosis. include rash and reversible increases in liver enzyme activities.
AMB cochleates are phosphatidylserine-calcium precipitates Flucytosine toxicity may result from conversion of flucytosine
in the form of a continuous lipid bilayer sheet rolled up in a to 5-fluorouracil by the intestinal microflora.
spiral. This formulation permits oral administration of AMB.
Cochleates have been effective when used to treat mouse mod- Griseofulvin
els of candidiasis and aspergillosis without significant toxicity.
They remain under investigation on a research basis. Mechanism of Action and Spectrum of Activity
Griseofulvin is an oral antifungal drug derived from Penicillium
5-Flucytosine griseofulvum that binds to fungal tubulin, leading to impaired
microtubule function and mitotic arrest. It also has antiinflam-
Mechanism of Action and Spectrum of Activity matory properties.42 After administration, griseofulvin is rap-
Flucytosine is a fluorinated pyrimidine related to fluorouracil. idly deposited in keratin precursor cells in the skin and hair, but
It has activity only against Cryptococcus spp. and Candida disappears from the stratum corneum within 48 to 72 hours of
spp. These fungi deaminate flucytosine to 5-fluorouracil, which discontinuation of treatment. Griseofulvin is fungistatic and has
interferes with DNA replication and protein synthesis. Mam- a limited spectrum of activity. It is used primarily to treat der-
malian cells cannot convert flucytosine into 5-fluorouracil, so matophytosis and is not effective against yeasts such as Candida
toxicity to mammalian cells is limited. Marked drug resistance spp. and Malassezia spp. Resistance has been documented in
arises during treatment (secondary drug resistance), and so flu- some dermatophytes and may be due to altered tubulin.
cytosine must always be used in combination with other drugs,
most commonly AMB. Flucytosine may be synergistic with Clinical Use
AMB. Resistance results from modifications in fungal enzymes The absorption of griseofulvin is increased when it is adminis-
that are required for flucytosine uptake and metabolism.37 tered with a fatty meal or whole milk. The drug is available in
microsized and ultramicrosized formulations, which improve
Clinical Use absorption from the stomach and small intestine. Most veterinary
Flucytosine is absorbed rapidly and well from the gastrointesti- preparations are microsized. The dose of the ultramicrosized for-
nal tract and is minimally bound to plasma proteins. Penetration mulation can be reduced by 50%, because of improved absorp-
of the CSF and aqueous humor is excellent. In cats, flucytosine tion, but efficacy remains unchanged (Table 9-4). Prolonged
may be considered to treat severe or refractory cryptococcosis treatment periods (months) are required for dermatophytosis (see
in combination with other agents. Because about 80% of the Chapter 58).43
dose is excreted unchanged in the urine, high urinary concentra-
tions can be achieved. For the same reason, flucytosine toxicity Adverse Effects
is greatly increased in animals with renal failure, so the drug Griseofulvin is a potent inducer of cytochrome P450 enzymes
should be avoided in these animals or the dose lowered and and so decreases the efficacy of other drugs that are metabolized
plasma drug concentrations monitored carefully (Table 9-3). to inactive metabolites by P450 enzymes. It is teratogenic, so
In human patients, toxicity is correlated with 2-hour postpill it should not be used to treat pregnant animals. Other adverse
concentrations greater than 100 mg/L, and concentrations of effects of griseofulvin include inappetence, vomiting, and diar-
30 to 80 mg/L are suggested for treatment of cryptococco- rhea. In cats, it can cause myelosuppression, which is generally
sis.15 Because AMB can cause impaired renal function, careful reversible on discontinuation of treatment, but irreversible pan-
monitoring for toxicity is indicated when flucytosine is used in cytopenia has been reported.44 Myelosuppression may be more
conjunction with AMB. The use of flucytosine in animals has likely to occur in cats with FIV infection.45 The CBC should be
mostly been limited by its high cost. monitored every 2 weeks during treatment.
94 SECTION 2  Antiinfective Therapy

TABLE 9-4
Suggested Griseofulvin and Terbinafine Doses for Dogs and Cats
Drug Dose (mg/kg) Interval (hours) Species Route Comments
Griseofulvin 25 12 D, C PO Administration with fatty food improves absorption.
(microsized) Dose may be increased to 50 mg/kg q12h for refrac-
tory infections. Avoid in cats with FIV infections.
Do not use in pregnancy. Drug interactions pos-
sible. Monitor CBC.
Griseofulvin 15 12 D, C PO Avoid in cats with FIV infections. Do not use in preg-
(ultramicrosized) nancy. Drug interactions possible. Monitor CBC.
Terbinafine 30 to 40 24 D, C PO Administer with food.
hydrochloride

C, Cats; D, dogs.

Terbinafine and the azoles. The prototype drug is caspofungin acetate (Can-
cidas). Other drugs in this class are micafungin and anidula-
Mechanism of Action and Spectrum of Activity fungin. Caspofungin is fungicidal against Candida spp. and
Terbinafine is a synthetic allylamine that inhibits fungal squa- fungistatic against Aspergillus species. Treatment with caspo-
lene epoxidase, which blocks fungal lanosterol and ergosterol fungin induced remission in a dog with disseminated aspergil-
synthesis and leads to accumulation of toxic squalene, with losis (1 mg/kg IV in 250 mL 0.9% NaCl over 1 hour, q24h).25
resultant fungal cell lysis. It is most effective for treatment of Synergism has been reported when echinocandins are used in
dermatophytosis. It has also been used to treat Malassezia spp. combination with other antifungal drugs.57 The echinocandins
dermatitis in dogs.46 The efficacy of terbinafine for treatment are ineffective against Cryptococcus spp., which possess little
of invasive fungal infections has not been well investigated. glucan synthase, and were only effective for treatment of Coc-
It has been used both successfully47 and unsuccessfully48 in cidioides spp. in a mouse model when used in conjunction with
combination with other drugs to treat pythiosis in dogs, and AMB.58 In human patients, caspofungin is approved for treat-
as an alternative to itraconazole to treat human sporotri- ment of refractory invasive aspergillosis, but it is also effective
chosis.49 It also has in vitro activity against other dimorphic for mucosal candidiasis.
fungi and molds, although its activity is highest for dermato- Caspofungin is given once daily as a slow IV infusion. In
phytes.50,51 Terbinafine may be synergistic when administered human patients, it is extensively protein bound and metabolized
with other antifungal drugs, which may provide enhanced slowly by the liver, with some renal excretion. It has limited
efficacy for treatment of refractory mycoses.52 However, non- ability to penetrate the CNS and eye.59,60 The cost is similar to
dermatophyte molds including Aspergillus spp. and Fusarium that of lipid formulations of amphotericin B. Adverse effects
spp. have been detected in people with onychomycosis that noted in humans include fever, phlebitis, and increased activity
failed to respond to terbinafine treatment.53 Resistance has of serum ALT (<20% of patients). The proper dose and extent
been reported in dermatophytes as a result of altered squalene to which adverse effects occur in dogs and cats is unknown, but
epoxidase.54 administration of anidulafungin to the dog treated with caspo-
fungin was associated with the development of a severe diffuse
Clinical Use urticarial reaction.25
Terbinafine is available in tablet form (Lamasil). It is well
absorbed in dogs when given with food (see Table 9-4).55 The Other Antifungal Treatments
drug is highly lipophilic and accumulates in skin, claws, hair,
and fat, where it persists for weeks after treatment is discontin- In human patients, hyperbaric oxygen therapy has been used
ued. Hepatic and renal failure increase plasma levels unpredict- to treat some fungal infections in combination with antifungal
ably, and dosage reduction may be required. drugs. Whether this is beneficial or harmful is controversial and
requires further study.61
Adverse Effects
Terbinafine is generally well tolerated. Gastrointestinal signs, SUGGESTED READINGS
especially vomiting and increased liver enzyme activities, are
uncommon. Facial pruritis has been reported in cats.56 Arthur RR, Drew RH, Perfect JR. Novel modes of antifungal drug
administration. Expert Opin Investig Drugs. 2004;13(8):903-932.
Goodwin ML, Drew RH. Antifungal serum concentration monitoring:
Echinocandins an update. J Antimicrob Chemother. 2008;61:17-25.
Hope WM, Billaud EM, Lestner J, et al. Therapeutic drug monitoring
The echinocandins are lipopeptide antifungals that inhibit the for triazoles. Curr Opin Infect Dis. 2008;21:580-586.
formation of β-1,3-d-glucans in the fungal cell wall, a mecha- Pound MW, Townsend ML, Dimondi V, et al. Overview of treatment
nism of action that is completely different from that for AMB options for invasive fungal infections. Med Mycol. 2011;49:561-580.
CHAPTER 9  Antifungal Drugs 95

REFERENCES 25. Schultz RM, Johnson EG, Wisner ER, et al. Clinicopathologic and
diagnostic imaging characteristics of systemic aspergillosis in 30
1. Sykes JE, Sturges BK, Cannon MS, et al. Clinical signs, imaging fea- dogs. J Vet Intern Med. 2008;22:851-859.
tures, neuropathology, and outcome in cats and dogs with central 26. Roffey SJ, Cole S, Comby P, et al. The disposition of voriconazole
nervous system cryptococcosis from California. J Vet Intern Med. in mouse, rat, rabbit, guinea pig, dog, and human. Drug Metab
2010;24:1427-1438. Dispos. 2003;31:731-741.
2. Clinical and Laboratory Standards Institute. M27–A3: Reference 27. Quimby JM, Hoffman SB, Duke J, et al. Adverse neurologic events
method for broth dilution antifungal susceptibility testing of yeasts. associated with voriconazole use in 3 cats. J Vet Intern Med.
2008. Available from www.clsi.org. 2010;24:647-649.
3. Clinical and Laboratory Standards Institute. M38–A2: Reference 28. Evans N, Gunew M, Marshall R, et al. Focal pulmonary granuloma
method for broth dilution antifungal susceptibility testing of fila- caused by Cladophialophora bantiana in a domestic short haired
mentous fungi. 2008. Available from www.clsi.org. cat. Med Mycol. 2011;49:194-197.
4. Negre A, Bensignor E, Guillot J. Evidence-based veterinary derma- 29. Wray JD, Sparkes AH, Johnson EM. Infection of the subcutis of the
tology: a systematic review of interventions for Malassezia derma- nose in a cat caused by Mucor species: successful treatment using
titis in dogs. Vet Dermatol. 2009;20:1-12. posaconazole. J Feline Med Surg. 2008;10:523-527.
5. Rex JH, Stevens DA. Systemic antifungal agents. In: Mandell GL, 30. McLellan GJ, Aquino SM, Mason DR, et al. Use of posaconazole
Bennett JE, Dolin R, eds. Mandell, Douglas and Bennett’s Prin- in the management of invasive orbital aspergillosis in a cat. J Am
ciples and Practice of Infectious Diseases. 7th ed. Philadelphia, PA: Anim Hosp Assoc. 2006;42:302-307.
Churchill Livingstone Elsevier; 2011:549-563. 31. Krishna G, Moton A, Ma L, et al. Pharmacokinetics and absorption
6. Mayer UK, Glos K, Schmid M, et al. Adverse effects of ketoconazole of posaconazole oral suspension under various gastric conditions in
in dogs—a retrospective study. Vet Dermatol. 2008;19:199-208. healthy volunteers. Antimicrob Agents Chemother. 2009;53:958-966.
7. Medleau L, Chalmers SA. Ketoconazole for treatment of dermato- 32. Li Y, Theuretzbacher U, Clancy CJ, et  al. Pharmacokinetic/phar-
phytosis in cats. J Am Vet Med Assoc. 1992;200:77-78. macodynamic profile of posaconazole. Clin Pharmacokinet.
8. Lien YH, Huang HP. Use of ketoconazole to treat dogs with pitu- 2010;49:379-396.
itary-dependent hyperadrenocorticism: 48 cases (1994-2007). J Am 33. Krockenberger MB, Martin P, Halliday C, et al. Localised Micros-
Vet Med Assoc. 2008;233:1896-1901. phaeropsis arundinis infection of the subcutis of a cat. J Feline Med
9. Mouatt JG. Cyclosporin and ketoconazole interaction for treat- Surg. 2010;12:231-236.
ment of perianal fistulas in the dog. Aust Vet J. 2002;80:207-211. 34. Thompson 3rd GR, Wiederhold NP. Isavuconazole: a comprehen-
10. Hugnet C, Lespine A, Alvinerie M. Multiple oral dosing of keto- sive review of spectrum of activity of a new triazole. Mycopatholo-
conazole increases dog exposure to ivermectin. J Pharm Pharm Sci. gia. 2010;170:291-313.
2007;10:311-318. 35. Oliva G, Roura X, Crotti A, et al. Guidelines for treatment of leish-
11. Pound MW, Townsend ML, Dimondi V, et  al. Overview of maniasis in dogs. J Am Vet Med Assoc. 2010;236:1192-1198.
treatment options for invasive fungal infections. Med Mycol. 36. Sundar S, Chakravarty J, Agarwal D, et al. Single-dose liposomal
2011;49:561-580. amphotericin B for visceral leishmaniasis in India. N Engl J Med.
12. Barone JA, Moskovitz BL, Guarnieri J, et  al. Enhanced bioavail- 2010;362:504-512.
ability of itraconazole in hydroxypropyl-beta-cyclodextrin solution 37. Espinel-Ingroff A. Mechanisms of resistance to antifungal agents:
versus capsules in healthy volunteers. Antimicrob Agents Che- yeasts and filamentous fungi. Rev Iberoam Micol. 2008;25:101-106.
mother. 1998;42:1862-1865. 38. Arthur RR, Drew RH, Perfect JR. Novel modes of antifungal drug
13. Legendre AM, Rohrbach BW, Toal RL, et  al. Treatment of administration. Expert Opin Investig Drugs. 2004;13:903-932.
blastomycosis with itraconazole in 112 dogs. J Vet Intern Med. 39. Malik R, Craig AJ, Wigney DI, et al. Combination chemotherapy
1996;10:365-371. of canine and feline cryptococcosis using subcutaneously adminis-
14. Pasqualotto AC, Denning DW. Generic substitution of itraconazole tered amphotericin B. Aust Vet J. 1996;73:124-128.
resulting in sub-therapeutic levels and resistance. Int J Antimicrob 40. Laniado-Laborin R, Cabrales-Vargas MN. Amphotericin B: side
Agents. 2007;30:93-94. effects and toxicity. Rev Iberoam Micol. 2009;26:223-227.
15. Goodwin ML, Drew RH. Antifungal serum concentration monitor- 41. Goldblum D, Rohrer K, Frueh BE, et  al. Ocular distribution of
ing: an update. J Antimicrob Chemother. 2008;61:17-25. intravenously administered lipid formulations of amphotericin
16. Hope WW, Billaud EM, Lestner J, et al. Therapeutic drug monitor- B in a rabbit model. Antimicrob Agents Chemother. 2002;46:
ing for triazoles. Curr Opin Infect Dis. 2008;21:580-586. 3719-3723.
17. Gupta AK, Chwetzoff E, Del Rosso J, et al. Hepatic safety of itra- 42. Moossavi M, Bagheri B, Scher RK. Systemic antifungal therapy.
conazole. J Cutan Med Surg. 2002;6:210-213. Dermatol Clin. 2001;19:35-52.
18. McKay JS, Cox CL, Foster AP. Cutaneous alternariosis in a cat. 43. Moriello KA. Treatment of dermatophytosis in dogs and cats:
J Small Anim Pract. 2001;42:75-78. review of published studies. Vet Dermatol. 2004;15:99-107.
19. Foy DS, Trepanier LA. Antifungal treatment of small animal veterinary 44. Rottman JB, English RV, Breitschwerdt EB, et  al. Bone marrow
patients. Vet Clin North Am Small Anim Pract. 2010;40:1171-1188. hypoplasia in a cat treated with griseofulvin. J Am Vet Med Assoc.
20. Ballent M, Lifschitz A, Virkel G, et  al. Modulation of the 1991;198:429-431.
P-glycoprotein-mediated intestinal secretion of ivermectin: in vitro 45. Shelton GH, Grant CK, Linenberger ML, et al. Severe neutropenia
and in vivo assessments. Drug Metab Dispos. 2006;34:457-463. associated with griseofulvin therapy in cats with feline immunode-
21. Bradbury BD, Jick SS. Itraconazole and fluconazole and certain ficiency virus infection. J Vet Intern Med. 1990;4:317-319.
rare, serious adverse events. Pharmacotherapy. 2002;22:697-700. 46. Rosales MS, Marsella R, Kunkle G, et al. Comparison of the clini-
22. Magrath GN, Pulido JS, Montero J, et  al. Cystoid macular cal efficacy of oral terbinafine and ketoconazole combined with
edema secondary to fluconazole toxicity. Ocul Immunol Inflamm. cephalexin in the treatment of Malassezia dermatitis in dogs—a
2010;18:472-474. pilot study. Vet Dermatol. 2005;16:171-176.
23. Lat A, Thompson 3rd GR. Update on the optimal use of vori- 47. Hummel J, Grooters A, Davidson G, et al. Successful management
conazole for invasive fungal infections. Infect Drug Resist. of gastrointestinal pythiosis in a dog using itraconazole, terbin-
2011;4:43-53. afine, and mefenoxam. Med Mycol. 2011;49:539-542.
24. Okabayashi K, Imaji M, Osumi T, et al. Antifungal activity of itra- 48. Pereira DI, Schild AL, Motta MA, et  al. Cutaneous and gastro­
conazole and voriconazole against clinical isolates obtained from ani- intestinal pythiosis in a dog in Brazil. Vet Res Commun. 2010;
mals with mycoses. Nihon Ishinkin Gakkai Zasshi. 2009;50:91-94. 34:301-306.
96 SECTION 2  Antiinfective Therapy

49. Francesconi G, Francesconi do Valle AC, Passos SL, et al. Compar- 55. Sakai MR, May ER, Imerman PM, et al. Terbinafine pharmacoki-
ative study of 250 mg/day terbinafine and 100 mg/day itraconazole netics after single dose oral administration in the dog. Vet Derma-
for the treatment of cutaneous sporotrichosis. Mycopathologia. tol. 2011;22:528-534.
2011;171:349-354. 56. Foust AL, Marsella R, Akucewich LH, et al. Evaluation of persis-
50. Ryder NS. Activity of terbinafine against serious fungal pathogens. tence of terbinafine in the hair of normal cats after 14 days of daily
Mycoses. 1999;42(Suppl 2):115-119. therapy. Vet Dermatol. 2007;18:246-251.
51. Gupta AK, Kohli Y, Batra R. In  vitro activities of posaconazole, 57. Chen SC, Slavin MA, Sorrell TC. Echinocandin antifungal drugs in
ravuconazole, terbinafine, itraconazole and fluconazole against fungal infections: a comparison. Drugs. 2011;71:11-41.
dermatophyte, yeast and non-dermatophyte species. Med Mycol. 58. Gónzalez GM, Gónzalez G, Najvar LK, et al. Therapeutic efficacy of
2005;43:179-185. caspofungin alone and in combination with amphotericin B deoxy-
52. Revankar SG, Nailor MD, Sobel JD. Use of terbinafine in rare and cholate for coccidioidomycosis in a mouse model. J Antimicrob
refractory mycoses. Future Microbiol. 2008;3:9-17. Chemother. 2007;60:1341-1346.
53. Baudraz-Rosselet F, Ruffieux C, Lurati M, et  al. Onychomycosis 59. Goldblum D, Fausch K, Frueh BE, et  al. Ocular penetration of
insensitive to systemic terbinafine and azole treatments reveals caspofungin in a rabbit uveitis model. Graefes Arch Clin Exp Oph-
non-dermatophyte moulds as infectious agents. Dermatology. thalmol. 2007;245:825-833.
2010;220:164-168. 60. Flattery AM, Hickey E, Gill CJ, et  al. Efficacy of caspofungin
54. Osborne CS, Leitner I, Hofbauer B, et al. Biological, biochemical, in a juvenile mouse model of central nervous system candidiasis.
and molecular characterization of a new clinical Trichophyton Antimicrob Agents Chemother. 2011;55:3491-3497.
rubrum isolate resistant to terbinafine. Antimicrob Agents Che- 61. Grahl N, Shepardson KM, Chung D, et  al. Hypoxia and fungal
mother. 2006;50:2234-2236. pathogenesis: to air or not to air? Eukaryot Cell. 2012;11:560-570.
CHAPTER 10

Antiprotozoal Drugs
Jane E. Sykes and Mark G. Papich

w KEY POINTS
• A  ntiprotozoal drugs often have a restricted spectrum of activ- • R  esistance to antiprotozoal drugs is a growing problem among
ity, although some are also active against bacteria and fungi. protozoal pathogens.
Many interfere with enzyme pathways specific to certain proto- • Some antiprotozoal drugs have minimal to no adverse effects,
zoal species. whereas for others, adverse effects severely limit use.
• Treatment with antiprotozoal drugs may not consistently clear • The availability of antiprotozoal drugs is restricted in some
an infection. countries.

INTRODUCTION can result from thiamine deficiency, which is reversible on addi-


tion of thiamine to the diet. However, thiamine supplementa-
The use of antiprotozoal drugs in dogs and cats is frequently tion may interfere with the drug’s efficacy.
extrapolated from their use in human patients or food animal spe-
cies. Almost all antiprotozoal drugs are not specifically approved Benzimidazoles
for treatment of protozoal infections in dogs and cats. Some Fenbendazole and Albendazole
antibacterial and antifungal drugs also have antiprotozoal activ- Benzimidazoles bind to β-tubulin within a variety of helminths
ity (see Chapters 8 and 9 for the use and adverse effects of these and protozoa. This leads to inhibition of tubulin polymerization
drugs). In vitro activity of antiprotozoal drugs, and monitoring of and the formation of microtubules, with impaired cell division.
resistance, is more difficult for antiprotozoal drugs because stan- Glucose uptake by parasites is also impaired. Resistance can
dardized susceptibility testing is not routinely performed for these result from production of altered β-tubulin by parasites, which
pathogens. In addition, many antiprotozoal drugs are designed to reduces binding of benzimidazole drugs.
be active in the lumen of the intestine for treatment of intestinal Fenbendazole is widely used to treat giardiasis in dogs and
protozoal infections and the concentration of active drug in the cats. It is safer than metronidazole, can be administered to young
intestinal lumen after oral administration is difficult to measure. animals, and has higher efficacy, although treatment failure can
Therefore, the concentration of drug to which these pathogens are still occur. A second course of treatment or administration of
exposed is often not known. The activity and dosage regimens of fenbendazole in combination with metronidazole can be effec-
antiprotozoal drugs are often based on the results of clinical trials, tive in refractory cases. Administration with food may improve
rather than concentration-exposure relationships between anti- absorption, but the fat content of the food does not influence
protozoal drugs and the organism of interest. absorption.1 Adverse effects of fenbendazole are very rare but
Many antiprotozoal drugs are active only against a restricted can include decreased appetite, vomiting, diarrhea, and rarely
range of protozoal species. To reflect this, drugs in this chapter are reversible pancytopenia.2 At high doses used to treat Mesoces-
organized into antiprotozoal drugs primarily used for gastroin- toides spp. peritonitis (100 mg/kg q12h), neurologic signs have
testinal infections; those with a broad spectrum of activity; those been observed.3 Febantel is metabolized to a benzimidazole
used to treat systemic protozoal infections (such as hepatozoono- compound and has been used in combination with praziquantel
sis, toxoplasmosis, neosporosis, and sarcocystosis); antiprotozoal and pyrantel (Drontal Plus) to treat Giardia spp. infection in
drugs used to treat leishmaniosis; and those used to treat Chagas’ dogs, although efficacy at label dosages has been variable and
disease. For many infections, treatment does not consistently some dogs can re-shed low numbers of cysts when treatment
result in parasitologic cure. is discontinued.4 Albendazole has an affinity for rapidly divid-
ing cells, and although it is used extensively for treatment of
parasitic infections in human patients, it been associated with
Antiprotozoal Drugs Used Primarily anorexia and reversible bone marrow suppression in dogs and
for Gastrointestinal Infections cats especially when high doses are administered for more than
5 days.5 As a result, fenbendazole is used more commonly in
Amprolium small animals.
Amprolium is a thiamine analogue that is used to prevent and
treat intestinal coccidiosis (Table 10-1). It is available as a feed Nitroimidazoles
additive for livestock and is sometimes administered in food Protozoa reduce nitroimidazoles to nitro anion free radicals,
or drinking water to puppies and kittens. Adverse effects of which cause damage to parasite DNA. Some nitroimidazoles are
anorexia or diarrhea are rare and primarily occur at high doses mutagens and carcinogens, but carcinogenesis has not been dem-
and with prolonged use. Central nervous system (CNS) signs onstrated in dogs and cats with long-term use. Metronidazole,

97
98 SECTION 2  Antiinfective Therapy

TABLE 10-1
Suggested Doses of Drugs That Are Primarily Used to Treat Protozoal Infections of the Gastrointestinal Tract in Small Animals
Dose Interval Duration
Drug (mg/kg) (hours) Species Route (days) Comments
Fenbendazole 50 24 C, D PO 5 Giardiasis. May be administered with
food. Safe in pregnancy.
Albendazole 25 12 C, D PO 3 Giardiasis. May cause bone marrow sup-
pression. Do not use in pregnancy.
Metronidazole 15 12 C, D PO 8 Giardiasis. Use caution with hepatic
insufficiency. Dose for metronidazole
benzoate is 25 mg/kg.
Tinidazole 15 12 D PO 5 Giardiasis. Administer with food or in
24 C capsules to reduce bitterness.
Ronidazole 30 24 C PO 14 Tritrichomoniasis. Avoid doses ≥60 mg/
kg/day. Compounded from powder.
Paromomycin 10 8 D PO 5-10 Cryptosporidiosis. Caution in animals
with diarrhea due to possible systemic
absorption. Avoid in cats.
Nitazoxanide 100 mg/animal 12 D, C PO 3 Cryptosporidiosis. Efficacy and safety
unclear. Vomiting common in cats.
Amprolium 1.25 g of 20% 24 D, C PO 7 Isosporiasis. Add to food. Do not ad-
powder or 30 mL minister for prolonged periods.
of 9.6% solution
to 3.8 L of water
Sulfadimethoxine 55 on day 1, 27 24 D, C PO 3-23 or for 48 hr Isosporiasis with or without a dihydro-
thereafter after signs resolve folate reductase inhibitor.

C, Cats; D, dogs.

ronidazole, and tinidazole have primarily been used to treat and tinidazole.8-10 Resistance to ronidazole has been identified
enteric protozoal infections. Benznidazole is specifically used to in some isolates of T. foetus and is associated with treatment
treat infections with Trypanosoma cruzi. failure in infected cats.11 Resistance is thought to result from
increased oxygen-scavenging capacity by the parasite, whereby
Metronidazole oxygen competes effectively with ronidazole and other nitro-
Metronidazole is used to treat giardiasis in dogs and cats, imidazoles for ferredoxin-bound electrons.
although efficacy may be as low as 50%. It also has activity Ronidazole is absorbed rapidly and completely after oral
against amoebic infections. The clinical use and adverse effects administration to cats. Some compounded formulations may
of metronidazole are described in Chapter 8. Doses of metro- have decreased efficacy as a result of low ronidazole content or
nidazole used for treatment of giardiasis have the potential to differences in drug release at the site of action (the large bowel).
be associated with neurotoxicity, so fenbendazole is preferred A modified-release formulation that is delivered to the colon
because of greater safety and efficacy. Metronidazole can be may have improved efficacy.12 Decreased appetite, vomiting,
combined with fenbendazole for refractory giardiasis. and neurologic signs can occur in dogs and cats, especially at
doses above 30 mg/kg q12h in cats and at doses as low as 10 mg/
Tinidazole kg/d in dogs.13 Once daily dosing is probably sufficient because
Tinidazole is a 5-nitroimidazole that has amoebicidal, giardicidal, of the long half-life of the drug in cats.12 Doses of 20 mg/kg or
trichomonicidal, and anaerobic bactericidal activity. It is some- less may not effectively clear infection with T. foetus. Neuro-
times used as a single-dose treatment for giardiasis in human logic signs result from γ-aminobutyric acid (GABA) antagonism
patients. The efficacy of tinidazole for treatment for giardiasis in in the CNS and include ataxia, decreased mentation, agitation,
dogs and cats has not been evaluated, and the half-life in dogs (4.4 tremors, and hyperesthesia, which occur up to 9 days after the
hours) and cats (8.4 hours) is shorter than that in human patients start of treatment and resolve when the drug is discontinued.13
(>12 hours).6,7 Tinidazole is very well absorbed in dogs and cats,
with a bioavailability of 100%. Adverse effects are similar to those Nitozoxanide
of metronidazole. Like metronidazole, tinidazole has a bitter taste. Nitazoxanide is a nitrothiazolyl-salicylamide derivative that has
activity against Giardia spp., Cryptosporidium spp., Sarcocystis
Ronidazole neurona, some anaerobic bacteria, Helicobacter spp., and Cam-
Ronidazole is the drug of choice for treatment of Tritrichomonas pylobacter jejuni. It inhibits the pyruvate-ferredoxin/flavodoxin
foetus infections, which are less responsive to metronidazole oxidoreductase enzyme-dependent electron transfer reaction
CHAPTER 10  Antiprotozoal Drugs 99

that is essential for anaerobic metabolism in these organisms. cats. Pyrimethamine is available as a single agent in tablets but
Resistance has been documented in Giardia spp.14 should be administered with a sulfonamide for the best efficacy.
Reports of nitazoxanide use in dogs and cats have been rare, Another alternative is the combination of pyrimethamine-sulfa-
and its efficacy in dogs and cats is largely unknown. An equine diazine, which is available in an oral liquid suspension for horses
formulation (Navigator) that was used to treat equine proto- (ReBalance). Although off-label, it is a convenient formulation
zoal meningoencephalitis caused by Sarcocystis neurona has for small animal veterinarians. This formulation can be admin-
been removed from the market. Doses have been extrapolated istered at a dose of 1 mg/kg pyrimethamine + 20 mg/kg sulfa-
from those used for human patients. Nitazoxanide treatment of diazine PO q24h. This is equivalent to 0.33 mL of the equine
cats co-infected with Cryptosporidium spp. and T. foetus led to formulation per 4 kg of body weight for dogs and cats.
cessation of shedding during treatment, but infection was not
eliminated.15 Vomiting occurred frequently, especially at higher Macrolides and Lincosamides
doses (75 mg/kg PO q12h). In humans, nitazoxanide is rapidly Clindamycin, azithromycin, and clarithromycin have antipro-
absorbed from the gastrointestinal tract and metabolized to the tozoal activity. The use of these macrolides and lincosamides in
active metabolite tizoxanide, which is highly protein bound. dogs and cats and their adverse effects are discussed in Chap-
After hepatic glucuronidation, it is excreted in urine and bile. ter 8. Clindamycin is the most widely used antiprotozoal for
treatment of toxoplasmosis and neosporosis in dogs and cats.
Although clindamycin inhibits shedding of Toxoplasma gondii
Antibacterial Drugs with Broad-Spectrum oocysts by cats,19 clinical efficacy of clindamycin for treating
Antiprotozoal Activity toxoplasmosis in dogs and cats has been questioned by experts
and in published studies. Trimethoprim-sulfonamides are a suit-
Folic Acid Antagonists able alternative, or if clindamycin is used, pyrimethamine may
Trimethoprim, pyrimethamine, ormetoprim, and sulfadiazine be used in combination. In human patients, pyrimethamine and
inhibit parasite replication through folate antagonism. Synergis- clindamycin are used as a substitute for pyrimethamine and
tic combinations of sulfadiazine with trimethoprim or pyrimeth- sulfadiazine for treatment of toxoplasmosis in sulfadiazine-
amine are primarily used to treat toxoplasmosis, neosporosis, sensitive individuals. Azithromycin is used in combination with
and intestinal coccidiosis (Isospora spp. infections) in dogs and atovaquone for treatment of babesiosis and cytauxzoonosis.
cats. A combination of pyrimethamine, trimethoprim-sulfadia-
zine, and clindamycin has also been used to treat Hepatozoon Paromomycin
americanum infections.16 The mechanisms of action, use, and Paromomycin is the only aminoglycoside antibiotic that has
adverse effects of trimethoprim and sulfonamides are discussed efficacy against protozoa. It is poorly absorbed from the gas-
in Chapter 8. trointestinal tract and so has been used to treat enteric proto-
zoal infections, particularly cryptosporidiosis. It is ineffective
Pyrimethamine for treatment of tritrichomoniasis in cats.10 Furthermore, when
Like trimethoprim, pyrimethamine inhibits dihydrofolate reduc- used to treat intestinal protozoal infections in cats, paromomy-
tase, which is necessary for synthesis of thymidine. However, in cin has been absorbed systemically because of intestinal muco-
contrast to trimethoprim, it has a greater affinity for the pro- sal compromise, with resultant acute renal failure, deafness,
tozoal enzyme than the bacterial enzyme. Resistance to pyri- and cataract formation.20 As a result, its use has been limited.
methamine can occur when parasites synthesize dihydrofolate In human patients, paromomycin has been used topically to
reductase enzymes with an altered drug target site. Pyrimeth- treat cutaneous leishmaniasis and parenterally to treat visceral
amine is well absorbed after oral administration and penetrates leishmaniasis.21
a variety of tissues including the CNS. Hepatic metabolism and
some renal excretion occur. Although clearance of pyrimeth- Tetracyclines and Ciprofloxacin
amine is not affected by renal disease, the use of caution with Doxycycline has primarily been used for malaria prophylaxis in
hepatic or renal insufficiency has been recommended in human humans. Ciprofloxacin is thought to inhibit DNA gyrase within
patients. a chloroplast organelle (the apicoplast) of apicomplexan para-
Pyrimethamine is well tolerated. Gastrointestinal signs such sites (see the triazines, later). It is an alternative to sulfadiazine
as vomiting, diarrhea, and decreased appetite occur in some for treatment of isosporiasis in human patients.18 Tetracyclines
treated animals. Bone marrow suppression can occur with pro- and ciprofloxacin have not been widely used for prevention or
longed treatment at higher doses as a result of folic acid defi- treatment of protozoal infections in dogs and cats with the pos-
ciency. In human patients, concurrent administration of folinic sible exception of doxycycline as part of combination treatment
acid is recommended when high doses are used for treatment for babesiosis (see Chapter 75).
of toxoplasmosis. Folinic acid, but not folic acid supplemen-
tation also reverses marrow suppression in dogs treated with
pyrimethamine.17 The CBC should be monitored weekly during Antiprotozoal Drugs Used for Systemic
treatment, and supplementation should be provided if leukope- Protozoal Infections
nia develops and continued treatment is necessary. Stomatitis,
ulcerative glossitis, and exfoliative dermatitis have also been Quinolone Derivatives
described in human patients as a result of folic acid deficiency.18 Atovaquone
Other adverse effects of pyrimethamine-sulfadiazine combina- Atovaquone is a hydroxynaphthoquinone that inhibits electron
tions result from the sulfadiazine component (see Chapter 8). transport in protozoa by targeting the cytochrome bc1 com-
Unlike trimethoprim-sulfadiazine, there are no approved plex (Table 10-2). It has been used in combination with other
formulations of pyrimethamine-sulfonamides for dogs and antiprotozoal drugs as an alternative treatment for malaria,
100 SECTION 2  Antiinfective Therapy

TABLE 10-2
Suggested Doses of Drugs Primarily Used to Treat Systemic Protozoal Diseases Excluding Leishmaniosis and Trypanosomiasis
in Small Animals
Dose Interval Duration
Drug (mg/kg) (hours) Species Route (days) Comments
Pyrimethamine 1 24 D PO 14-28 Primarily neosporosis, toxoplasmosis, and American hep-
0.5-1 C atozoonosis. Use with a sulfonamide. Use caution with
hepatic and renal insufficiency. Monitor CBC. Folinic
acid supplementation (5 mg/day) may be required.
Clindamycin 22 12 D, C PO Toxoplasmosis, neosporosis, sarcocystosis, and
Americ­an hepatozoonosis.
Azithromycin 10 24 D, C PO 10 Babesiosis and cytauxzoonosis. Used with atovaquone.
Atovaquone 13.3 8 D PO 10 Babesiosis and cytauxzoonosis with azithromycin.
15 C Administer with food.
Decoquinate 10-20 12 D PO ≥365 American hepatozoonosis and sarcocystosis. Powder
(6% decoquinate; 60 mg active ingredient per gram)
is mixed with food. This equates to 0.5 to 1 table-
spoon/10 kg body weight q12h.
Imidocarb 6.6 Once, D Deep IM N/A Large Babesia spp. infections. Caution with hepatic or
dipropionate 5 repeat in C renal insufficiency. Avoid use with other cholinesteras­e
14 days inhibitors.
Diminazene 3-5 Once D Deep IM N/A Babesiosis and African trypanosomiasis. Narrow
aceturate therapeutic range.
Ponazuril 20-50 12-24 D PO 3-28 Toxoplasmosis, neosporosis, isosporiasis. Optimal dose,
duration, efficacy, and adverse effects unknown.
Toltrazuril 5-10 12-24 D PO 1-14 Hepatozoonosis, isosporiasis. One dose may be effec-
18 C tive for isosporiasis. Optimal dose and duration for
hepatozoonosis unknown.

C, Cats; D, dogs; N/A, not applicable.

toxoplasmosis, and pneumocystosis in human patients.18 In result in cross-resistance to decoquinate.25 Although developed
veterinary medicine, atovaquone is used in combination with as a food additive for use in production animals, decoquinate
azithromycin for treatment of Babesia gibsoni and Babesia can be used successfully and without adverse effects to prevent
conradae infections and cytauxzoonosis (see Chapters 75 and relapses in dogs that are chronically infected with Hepatozoon
76), but is expensive. Resistance has been reported in Plas- americanum.16 Decoquinate also appeared to be effective for
modium spp., T. gondii, Pneumocystis jirovecii, and canine treatment of Sarcocystis spp. myositis in a dog.26 Its distribution
B. gibsoni strains as a result of mutations in the cytochrome and metabolism in dogs and cats have not been described.
bc1 complex.
Atovaquone is highly lipophilic and extensively protein Aromatic Diamines
bound. Little is known about atovaquone metabolism in dogs The aromatic diamines include imidocarb dipropionate, dim-
and cats. Its oral bioavailability increases significantly when food inazene aceturate, pentamidine isethionate, and phenamidine
is administered concurrently. Atovaquone is available alone or isethionate. These drugs inhibit DNA synthesis in protozoa. In
in a formulation with proguanil for treatment and prevention of dogs, imidocarb and diminazene have been used most widely,
malaria in human patients. The combination formulation may primarily for treatment of Babesia spp. and Hepatozoon canis
cause vomiting and diarrhea in dogs, but otherwise atovaquone infections. In general, the use of imidocarb has been preferred
appears well tolerated by both dogs and cats.22-24 In human over diminazene because of diminazene’s narrow therapeu-
patients, adverse effects have included gastrointestinal signs, tic index. Pentamidine and phenamidine have also been used
headache, fever, and increased liver enzyme activities. Coadmin- in dogs. Pentamidine isethionate is a second-line treatment for
istration with metoclopramide, tetracycline, or rifampin signifi- leishmaniasis in human patients and is used as an alternative
cantly decreases plasma drug concentrations in humans. to diminazene for treatment of African trypanosomiasis (see
­Chapter 78). Treatment of protozoal infections using the aro-
Decoquinate matic diamines may not result in complete parasite elimination.
Decoquinate is a 4-hydroxyquinolone coccidiostat that inhib-
its electron transport in protozoal mitochondria and interferes Imidocarb Dipropionate
with sporozoite development. It likely has a similar mechanism Imidocarb dipropionate is primarily used to treat large Babesia
of action to atovaquone, because resistance to atovaquone can spp. and Hepatozoon canis infections in dogs. More effective
CHAPTER 10  Antiprotozoal Drugs 101

agents have replaced imidocarb for treatment of canine mono- Dogs, Bayer Animal Health) for treatment of isosporiasis and
cytic ehrlichiosis and feline cytauxzoonosis. roundworm infections in puppies over 2 weeks of age. The sus-
Imidocarb is given by intramuscular injection, twice, 2 weeks pension contains 18 mg/mL of toltrazuril, and a single 9-mg/kg
apart. It is generally well tolerated, although it can cause tran- dose is recommended. Toltrazuril appeared to be as effective
sient pain at the site of injection. It appears to be eliminated as trimethoprim-sulfadiazine-clindamycin-pyrimethamine for
in urine and feces.27 Acute adverse effects result from its anti- treatment of canine American hepatozoonosis.16 One dog with
cholinergic activity and include vomiting, shivering, hypersali- Hepatozoon canis infection recovered after treatment with tol-
vation, lacrimation, diarrhea, agitation, lethargy, pyrexia, and trazuril and trimethoprim-sulfamethoxazole,42 but a toltrazuril
periorbital swelling. These generally resolve within a few hours. and imidocarb combination did not offer benefit over treat-
A possible association between imidocarb treatment and acute ment of H. canis infection with imidocarb alone.43 Dogs with
renal tubular necrosis has been reported in dogs.28 Massive H. canis infection respond clinically within 72 hours of starting
hepatic necrosis was described after overdosage of dogs with 10 treatment with toltrazuril. Anecdotal reports exist of the use of
times the recommended dose.29 ponazuril to treat Isospora spp. infections in dogs and cats, but
its efficacy is unclear. Doses of ponazuril used have ranged from
Diminazene Aceturate 20 to 50 mg/kg for 2 to 5 days. In one dog, ocular toxoplasmo-
Diminazene aceturate is administered parenterally, primarily to sis that was refractory to clindamycin resolved after treatment
dogs, for treatment of babesiosis, African trypanosomiasis, and, with ponazuril for 28 days.44
most recently, infections with Rangelia vitalii, a novel protozoal The pharmacokinetics of the triazines in dogs has not been
pathogen of dogs from Brazil.30 Diminazene aceturate is not reported. In horses, ponazuril crosses the blood-brain barrier to
readily available in the United States. Resistance to diminazene some extent and achieves concentrations in the cerebrospinal
has been described in Babesia gibsoni.31 fluid sufficient to inhibit protozoa. It has long half-life in horses
Although formulations for small animals are not available, (>4 days) and in cattle (2 to 3 days). Because of its specific activ-
a powdered commercial drug formulation (Veriben) has been ity against apicomplexan parasites, significant toxicity in mam-
reconstituted with sterile water to a concentration of 7 mg/mL malian species has not been reported.
and administered intramuscularly at a dose of 3 mg/kg dim-
inazene diaceturate for a pharmacokinetic study in cats.32 The Antileishmanial Antiprotozoal Drugs
dose was well tolerated, and diminazene was eliminated with
a half-life of only 1.7 hours and a peak concentration of only Antifungal Agents
0.5 µg/mL. Development of clinically effective doses from these Amphotericin B
data requires further studies. Diminazene was administered to Amphotericin B is a treatment of choice for human visceral leish-
seven cats experimentally infected with Trypanosoma evansi at maniasis.18 It is thought to bind to ergosterol in the protozoal
a dose of 3.5 mg/kg intramuscularly on five consecutive days.33 membrane, and blocks the ability of Leishmania spp. to bind to
The treatment was 85.7% efficacious for elimination of the par- and enter macrophages.45 Lipid amphotericin B formulations have
asite, and no adverse effects were observed. been used to treat visceral leishmaniosis in dogs, although organ-
Effective doses of diminazene approach doses that are toxic, ism persistence and relapse have been reported in some dogs after
so it should be used with caution.34 Adverse effects include twice-weekly treatment for up to 10 administrations (0.8 to 3.3 mg/
tachycardia and CNS signs such as ataxia, nystagmus, and opis- kg intravenously).46,47 In an effort to reduce selection for resistant
thotonos. A single treatment can be given, or the dose can be parasites, the World Health Organization recommends against the
repeated 72 to 96 hours after drug administration. In some pro- use of amphotericin B for treatment of canine leishmaniosis.48 A
tocols for B. gibsoni infections, diminazene administration has full discussion of amphotericin B is provided in Chapter 9.
been followed a day later by treatment with imidocarb.
Azole Antifungals
Triazine Antiprotozoals (Toltrazuril and Ponazuril) Azole antifungals such as fluconazole have been used to treat
Toltrazuril and its major metabolite ponazuril (toltrazuril sul- cutaneous leishmaniasis in human patients. More effective
fone, Marquis) are triazine-based antiprotozoal drugs that have antiprotozoal drugs are substituted for visceral leishmaniasis.
specific activity against apicomplexan coccidial infections. Tol- Posaconazole and ravuconazole also have activity against Try-
trazuril is not available within the United States. Ponazuril and panosoma cruzi (see Chapter 78), and fluconazole may have
toltrazuril appear to act on a plastid-like chloroplast organelle activity against Toxoplasma (see Chapter 72).
(the apicoplast) in apicomplexan protozoa, which may have
originally been acquired from a green alga.35 This organelle Antimony Compounds
contains a small circular genome and operates key biochemi- Sodium stibogluconate (Pentostam) and N-methylglucamine
cal pathways. Although their mechanism of action is unclear, antimoniate (meglumine antimoniate, Glucantime) are derived
triazines may inhibit metabolic enzymes or decrease pyrimi- from the heavy metal antimony (Sb). They are called pentava-
dine synthesis within the apicoplast. Ponazuril and toltrazuril lent antimony compounds (symbol Sbv) because they contain
have been used in animals to treat isosporiasis, toxoplasmosis, Sb atoms that have five electrons in their outer shell. Pentava-
neosporosis, and equine protozoal meningoencephalitis. Tri- lent antimonials have been recommended as the first choice for
azine resistance has been described in coccidia from production treatment of Leishmania infections in humans and in dogs.49
animals.36 Their mechanism of action is still not completely clear, but it
In dogs and cats, toltrazuril has been used to treat isospo- is thought that pentavalent antimony undergoes reduction to
riasis and hepatozoonosis, although infection may persist in the more toxic trivalent version, possibly within macrophage
some animals.16,37-42 In Europe it is available in combination phagolysosomes or within the parasite itself.50 The trivalent
with the anthelmintic emodepside (Procox Oral Suspension for compound inhibits protozoal enzymes and damages protozoal
102 SECTION 2  Antiinfective Therapy

TABLE 10-3
Suggested Doses of Drugs Primarily Used to Treat Leishmaniosis or Chagas’ Disease in Dogs
Dose Interval Duration
Drug (mg/kg) (hours) Route (days) Comments
Meglumine 75-100 24 SC 30-60 Leishmaniosis. Use with allopurinol. Available in ampules. Dose
antimoniate based on concentration of meglumine antimoniate. Caution
in dogs with renal insufficiency.
Allopurinol 10 12 PO At least Leishmaniosis. Use with meglumine or miltefosine. Monitor
6-12 months CBC and serum chemistry in dogs with hepatic and renal
insufficiency.
Miltefosine 2 24 PO 28 Leishmaniosis. Use with allopurinol.
Benznidazole 5-10 24 PO 60 Chagas’ disease

DNA.51 Resistance to antimonials is an emerging problem in more likely to occur in human patients with renal insufficiency. At
human Leishmania spp. isolates. This has led to the use of usual doses, allopurinol treatment leads to improvement in kidney
higher drug dosages, with an associated rise in the rate of drug lesions in dogs with leishmaniosis that have renal insufficiency.60
toxicity. The availability of antimonial drugs is limited in the Nevertheless, careful monitoring for adverse effects is indicated in
United States. these dogs (see Table 10-3). Serious drug interactions can occur
Both sodium stibogluconate and meglumine antimoniate are when allopurinol is administered with azathioprine.
administered subcutaneously (Table 10-3). The treatment of
choice for canine leishmaniosis is a combination of meglumine Miltefosine
antimoniate and allopurinol.48,49 Complete clearance of the infec- Miltefosine is an effective alternative to pentavalent antimonials
tion may not always occur.52 After subcutaneous administration, for treatment of leishmaniosis.61 It is a phospholipid analogue
more than 80% of meglumine antimoniate is eliminated by the that activates cellular proteases in Leishmania spp., which results
kidneys. Pain at the site of injection is the most common adverse in apoptosis. Miltefosine is the first highly active oral drug for
effect, and cutaneous abscesses or cellulitis may also occur.53 treatment of leishmaniosis. Nevertheless, despite clinical improve-
Systemic adverse effects such as lethargy, vomiting, diarrhea, ment in treated dogs, elimination of the parasite may not always
inappetence, and increased serum liver enzyme activities may be occur.62,63 Resistance to miltefosine can result from increased
more likely to occur in the presence of renal failure, which can be P-glycoprotein–mediated drug efflux and decreased drug uptake.
a complication of leishmaniosis. Of concern, the administration Because miltefosine has a long half-life and must be administered
of meglumine antimoniate to healthy dogs has led to renal tubu- for 28 days, selection for resistant isolates has been a concern.
lar necrosis, in the absence of azotemia.54 In human patients, The World Health Organization has recommended that veteri-
other adverse effects include generalized arthralgias, abdominal nary use of miltefosine in dogs with leishmaniosis be avoided in
pain, mild cytopenias, cardiotoxicity, and chemical pancreati- order to minimize the rate of selection for resistant strains.64
tis.21 Novel formulations of antimony compounds are being Over 90% of miltefosine is absorbed from the gastrointestinal
developed for treatment of leishmaniasis, including lipid formu- tract of dogs after oral administration.54 The half-life of miltefo-
lations and preparations that could be administered orally.50 sine in the dog is approximately 6 days, so it accumulates until a
steady-state plasma concentration is reached after 3 to 4 weeks
Allopurinol of treatment. Clearance is believed to result from slow hepatic
Allopurinol is a purine analogue. In parasites, allopurinol is metab- metabolism and excretion in bile. In dogs, adverse effects of treat-
olized to derivatives that are incorporated into RNA, which leads ment are mild and transient and consist of occasional vomiting
to impaired protein synthesis. Resistance can occur and appears to and diarrhea.65 In contrast to meglumine antimoniate, miltefos-
result from reduced activity of purine transporters and a reduced ine does not appear to contribute to renal pathology in dogs.54,66
ability to accumulate purine.55 Although it has been used alone for
treatment of canine leishmaniosis, allopurinol is most commonly Antiprotozoal Drugs for Treatment
used in combination with meglumine or miltefosine. of Chagas’ Disease
Allopurinol is rapidly absorbed from the gastrointestinal tracts Benznidazole
of dogs. Peak drug concentrations occur 1 to 3 hours after adminis- Benznidazole is a nitroimidazole that is used to treat acute Cha-
tration.56 Allopurinol is not bound to plasma proteins. Allopurinol gas’ disease (see Chapter 78). Benznidazole is activated by a
rarely causes adverse effects in dogs. It inhibits mammalian xan- parasite-specific nitroreductase to produce toxic metabolites.67
thine oxidase, which results in decreased uric acid production from Resistance can result from reduction in the level of activity of
xanthine. As a result, long-term use can result in xanthine urolithi- the nitroreductase and confers cross-resistance to nifurtimox.
asis.57 In human patients, allopurinol most commonly causes skin Benznidazole is lipophilic, is readily absorbed from the gas-
rashes. Gastrointestinal signs can also occur. Rarely administra- trointestinal tract, and undergoes hepatic metabolism. In the
tion of allopurinol to humans results in aplastic anemia or a hyper- United States, it is available from the Centers for Disease Con-
sensitivity syndrome characterized by toxic epidermal necrolysis trol and Prevention (CDC). The main adverse effect of treat-
with hepatic and renal failure.58,59 Serious adverse reactions are ment in dogs is vomiting. In human patients, it can cause skin
CHAPTER 10  Antiprotozoal Drugs 103

rashes, peripheral neuropathy, and less commonly bone mar- 17. Castles TR, Kintner LD, Lee CC. The effects of folic or folinic acid
row suppression. It also has the potential to be carcinogenic. on the toxicity of pyrimethamine in dogs. Toxicol Appl Pharmacol.
1971;20:447-459.
Nifurtimox 18. Moore TA. Agents active against parasites and Pneumocystis. In:
Nifurtimox is a nitrofuran derivative used to treat Chagas’ dis- Mandell GL, Bennett JE, Dolin R, eds. Mandell, Douglas and Ben-
nett’s Principles and Practice of Infectious Diseases. 7th ed. Phila-
ease in humans. It has limited efficacy, with 70% parasitologic
delphia, PA: Churchill Livingstone Elsevier; 2011:631-668.
cure for acute disease and at best 20% for chronic disease.18
19. Malmasi A, Mosallanejad B, Mohebali M, et  al. Prevention of
As with benznidazole, metabolic reduction of the nitro group shedding and reshedding of Toxoplasma gondii oocysts in experi-
by a trypanosome-specific nitroreductase leads to the forma- mentally infected casts treated with oral clindamycin: a preliminary
tion of toxic metabolites. In human patients, it is well absorbed study. Zoonoses Public Health. 2009;56(2):102-104.
orally and undergoes hepatic metabolism. Gastrointestinal and 20. Gookin JL, Riviere JE, Gilger BC, et al. Acute renal failure in four
neurologic signs are the primary adverse effects of nifurtimox, cats treated with paromomycin. J Am Vet Med Assoc. 1999;215:
and like benznidazole, it has carcinogenic properties.68 Severe 1821-1823:1806.
adverse effects have precluded the use of nifurtimox in dogs.69 21. Murray HW, Berman JD, Davies CR, et al. Advances in leishmani-
asis. Lancet. 2005;366:1561-1577.
SUGGESTED READING 22. Birkenheuer AJ, Levy MG, Breitschwerdt EB. Efficacy of com-
bined atovaquone and azithromycin for therapy of chronic Babe-
Rossignol JF. Cryptosporidium and Giardia: treatment options and
sia gibsoni (Asian genotype) infections in dogs. J Vet Intern Med.
prospects for new drugs. Exp Parasitol. 2010;124(1):45-53.
2004;18:494-498.
23. Cohn LA, Birkenheuer AJ, Brunker JD, et  al. Efficacy of atova-
REFERENCES quone and azithromycin or imidocarb dipropionate in cats with
1. McKellar QA, Galbraith EA, Baxter P. Oral absorption and bio- acute cytauxzoonosis. J Vet Intern Med. 2011;25:55-60.
availability of fenbendazole in the dog and the effect of concurrent 24. Matsuu A, Koshida Y, Kawahara M, et  al. Efficacy of atova-
ingestion of food. J Vet Pharmacol Ther. 1993;16:189-198. quone against Babesia gibsoni in vivo and in vitro. Vet Parasitol.
2. Gary AT, Kerl ME, Wiedmeyer CE, et al. Bone marrow hypoplasia 2004;124:9-18.
associated with fenbendazole administration in a dog. J Am Anim 25. Pfefferkorn ER, Borotz SE, Nothnagel RF. Mutants of Toxoplasma
Hosp Assoc. 2004;40:224-229. gondii resistant to atovaquone (566C80) or decoquinate. J Parasi-
3. Boyce W, Shender L, Schultz L, et  al. Survival analysis of dogs tol. 1993;79:559-564.
diagnosed with canine peritoneal larval cestodiasis (Mesocestoides 26. Sykes JE, Dubey JP, Lindsay LL, et  al. Severe myositis associ-
spp.). Vet Parasitol. 2011;180:256-261. ated with Sarcocystis spp. infection in 2 dogs. J Vet Intern Med.
4. Bowman DD, Liotta JL, Ulrich M, et  al. Treatment of naturally 2011;25(6):1277-1283.
occurring, asymptomatic Giardia sp. in dogs with Drontal Plus fla- 27. Vial HJ, Gorenflot A. Chemotherapy against babesiosis. Vet Para-
vour tablets. Parasitol Res. 2009;105(Suppl 1):S125-S134. sitol. 2006;138:147-160.
5. Stokol T, Randolph JF, Nachbar S, et  al. Development of bone 28. Máthé A, Dobos-Kovács M, Vörös K. Histological and ultrastruc-
marrow toxicosis after albendazole administration in a dog and tural studies of renal lesions in Babesia canis infected dogs treated
cat. J Am Vet Med Assoc. 1997;210:1753-1756. with imidocarb. Acta Vet Hung. 2007;55:511-523.
6. Lamp KC, Freeman CD, Klutman NE, et  al. Pharmacokinetics 29. Kock N, Kelly P. Massive hepatic necrosis associated with acci-
and pharmacodynamics of the nitroimidazole antimicrobials. Clin dental imidocarb dipropionate toxicosis in a dog. J Comp Pathol.
Pharmacokinet. 1999;36:353-373. 1991;104:113-116.
7. Sarkiala E, Järvinen A, Välttilä S, et al. Pharmacokinetics of tinida- 30. Da Silva AS, Franca RT, Costa MM, et al. Experimental infection
zole in dogs and cats. J Vet Pharmacol Ther. 1991;14:257-262. with Rangelia vitalii in dogs: acute phase, parasitemia, biological
8. Gookin JL, Copple CN, Papich MG, et al. Efficacy of ronidazole cycle, clinical-pathological aspects and treatment. Exp Parasitol.
for treatment of feline Tritrichomonas foetus infection. J Vet Intern 2011;128:347-352.
Med. 2006;20:536-543. 31. Hwang SJ, Yamasaki M, Nakamura K, et  al. Development and
9. Gookin JL, Stauffer SH, Coccaro MR, et al. Efficacy of tinidazole characterization of a strain of Babesia gibsoni resistant to diminaz-
for treatment of cats experimentally infected with Tritrichomonas ene aceturate in vitro. J Vet Med Sci. 2010;72:765-771.
foetus. Am J Vet Res. 2007;68:1085-1088. 32. Lewis KM, Cohn LA, Birkenheuer AJ, et  al. Pharmacokinetics
10. Kather EJ, Marks SL, Kass PH. Determination of the in  vitro of diminazene diaceturate in healthy cats. J Vet Pharmacol Ther.
susceptibility of feline Tritrichomonas foetus to 5 antimicrobial 2012;35:608-610.
agents. J Vet Intern Med. 2007;21:966-970. 33. Silva ASD, Zanette RA, Wolkmer P, et al. Diminazene aceturate in
11. Gookin JL, Stauffer SH, Dybas D, et al. Documentation of in vivo the control of Trypanosoma evansi infection in cats. Vet Parasitol.
and in vitro aerobic resistance of feline Tritrichomonas foetus iso- 2009;165:47-50.
lates to ronidazole. J Vet Intern Med. 2010;24:1003-1007. 34. Solano-Gallego L, Baneth G. Babesiosis in dogs and cats—
12. LeVine DN, Papich MG, Gookin JL, et al. Ronidazole pharmacoki- expanding parasitological and clinical spectra. Vet Parasitol.
netics after intravenous and oral immediate-release capsule admin- 2011;181:48-60.
istration in healthy cats. J Feline Med Surg. 2011;13:244-250. 35. Wiesner J, Reichenberg A, Heinrich S, et al. The plastid-like organ-
13. Rosado TW, Specht A, Marks SL. Neurotoxicosis in 4 cats receiv- elle of apicomplexan parasites as drug target. Curr Pharm Des.
ing ronidazole. J Vet Intern Med. 2007;21:328-331. 2008;14:855-871.
14. Müller J, Sterk M, Hemphill A, et al. Characterization of Giardia 36. Stephen B, Rommel M, Daugschies A, et al. Studies of resistance to
lamblia WB C6 clones resistant to nitazoxanide and to metronida- anticoccidials in Eimeria field isolates and pure Eimeria strains. Vet
zole. J Antimicrob Chemother. 2007;60:280-287. Parasitol. 1997;69:19-29.
15. Gookin JL, Levy MG, Law JM, et al. Experimental infection of cats 37. Altreuther G, Gasda N, Adler K, et  al. Field evaluations of the
with Tritrichomonas foetus. Am J Vet Res. 2001;62:1690-1697. ­efficacy and safety of emodepside plus toltrazuril (Procox oral
16. Macintire DK, Vincent-Johnson NA, Kane CW, et  al. Treatment suspension for dogs) against naturally acquired nematode and
of dogs infected with Hepatozoon americanum: 53 cases (1989- Isospora spp. infections in dogs. Parasitol Res. 2011;109(Suppl
1998). J Am Vet Med Assoc. 2001;218:77-82. 1):S21-S28.
104 SECTION 2  Antiinfective Therapy

38. Altreuther G, Gasda N, Schroeder I, et  al. Efficacy of emodep- 55. Kerby BR, Detke S. Reduced purine accumulation is encoded on an
side plus toltrazuril suspension (Procox oral suspension for dogs) amplified DNA in Leishmania mexicana amazonensis resistant to
against prepatent and patent infection with Isospora canis and toxic nucleosides. Mol Biochem Parasitol. 1993;60:171-185.
Isospora ohioensis-complex in dogs. Parasitol Res. 2011;109(Suppl 56. Ling GV, Case LC, Nelson H, et al. Pharmacokinetics of allopuri-
1):S9-20. nol in Dalmatian dogs. J Vet Pharmacol Ther. 1997;20:134-138.
39. Daugschies A, Mundt HC, Letkova V. Toltrazuril treatment of 57. Ling GV, Ruby AL, Harrold DR, et  al. Xanthine-containing
cystoisosporosis in dogs under experimental and field conditions. urinary calculi in dogs given allopurinol. J Am Vet Med Assoc.
Parasitol Res. 2000;86:797-799. 1991;198:1935-1940.
40. Lloyd S, Smith J. Activity of toltrazuril and diclazuril against Isos- 58. Kim YW, Park BS, Ryu CH, et  al. Allopurinol-induced aplastic
pora species in kittens and puppies. Vet Rec. 2001;148:509-511. anemia in a patient with chronic kidney disease. Clin Nephrol.
41. Petry G, Kruedewagen E, Bach T, et  al. Efficacy of Procox oral 2009;71:203-206.
suspension for dogs (0.1% emodepside and 2% toltrazuril) against 59. Fagugli RM, Gentile G, Ferrara G, et al. Acute renal and hepatic
experimental nematode (Toxocara cati and Ancylostoma tubae- failure associated with allopurinol treatment. Clin Nephrol.
forme) infections in cats. Parasitol Res. 2011;109(Suppl 1):S37-S43. 2008;70:523-526.
42. Voyvoda H, Pasa S, Uner A. Clinical Hepatozoon canis infection in 60. Plevraki K, Koutinas AF, Kaldrymidou H, et  al. Effects of allo-
a dog in Turkey. J Small Anim Pract. 2004;45:613-617. purinol treatment on the progression of chronic nephritis in
43. Pasa S, Voyvoda H, Karagenc T, et al. Failure of combination ther- canine leishmaniosis (Leishmania infantum). J Vet Intern Med.
apy with imidocarb dipropionate and toltrazuril to clear Hepato- 2006;20:228-233.
zoon canis infection in dogs. Parasitol Res. 2011;109:919-926. 61. Miró G, Oliva G, Cruz I, et  al. Multicentric, controlled clinical
44. Swinger RL, Schmidt Jr KA, Dubielzig RR. Keratoconjunctivitis study to evaluate effectiveness and safety of miltefosine and allopu-
associated with Toxoplasma gondii in a dog. Vet Ophthalmol. rinol for canine leishmaniosis. Vet Dermatol. 2009;20:397-404.
2009;12:56-60. 62. Manna L, Vitale F, Reale S, et al. Study of efficacy of miltefosine and
45. Paila YD, Saha B, Chattopadhyay A. Amphotericin B inhibits entry allopurinol in dogs with leishmaniosis. Vet J. 2009;182:441-445.
of Leishmania donovani into primary macrophages. Biochem Bio- 63. Andrade HM, Toledo VP, Pinheiro MB, et al. Evaluation of milte-
phys Res Commun. 2010;399:429-433. fosine for the treatment of dogs naturally infected with L. infantum
46. Oliva G, Gradoni L, Ciaramella P, et  al. Activity of liposomal (=L. chagasi) in Brazil. Vet Parasitol. 2011;181:83-90.
amphotericin B (AmBisome) in dogs naturally infected with Leish- 64. World Health Organization. Report of a WHO informal consul-
mania infantum. J Antimicrob Chemother. 1995;36:1013-1019. tation on liposomal amphotericin B in the treatment of visceral
47. Cortadellas O. Initial and long-term efficacy of a lipid emulsion of leishmaniosis. 2007. http://www.who.int/entity/neglected_diseases/
amphotericin B desoxycholate in the management of canine leish- resources/AmBisomeReport.pdf: Last accessed January 25, 2013.
maniasis. J Vet Intern Med. 2003;17:808-812. 65. Woerly V, Maynard L, Sanquer A, et al. Clinical efficacy and toler-
48. Solano-Gallego L, Koutinas A, Miro G, et  al. Directions for the ance of miltefosine in the treatment of canine leishmaniosis. Parasi-
diagnosis, clinical staging, treatment and prevention of canine tol Res. 2009;105:463-469.
leishmaniosis. Vet Parasitol. 2009;165:1-18. 66. Mateo M, Maynard L, Vischer C, et al. Comparative study on the
49. Oliva G, Roura X, Crotti A, et al. Guidelines for treatment of leish- short term efficacy and adverse effects of miltefosine and meglu-
maniasis in dogs. J Am Vet Med Assoc. 2010;236:1192-1198. mine antimoniate in dogs with natural leishmaniosis. Parasitol Res.
50. Frézard F, Demicheli C, Ribeiro RR. Pentavalent antimonials: new 2009;105:155-162.
perspectives for old drugs. Molecules. 2009;14:2317-2336. 67. Wilkinson SR, Taylor MC, Horn D, et al. A mechanism for cross-
51. Lima MI, Arruda VO, Alves EV, et  al. Genotoxic effects of the resistance to nifurtimox and benznidazole in trypanosomes. Proc
antileishmanial drug Glucantime. Arch Toxicol. 2010;84:227-232. Natl Acad Sci U S A. 2008;105:5022-5027.
52. Manna L, Reale S, Vitale F, et al. Real-time PCR assay in Leishma- 68. Castro JA, de Mecca MM, Bartel LC. Toxic side effects of drugs
nia-infected dogs treated with meglumine antimoniate and allopu- used to treat Chagas’ disease (American trypanosomiasis). Hum
rinol. Vet J. 2008;177:279-282. Exp Toxicol. 2006;25:471-479.
53. Ikeda-Garcia FA, Lopes RS, Ciarlini PC, et al. Evaluation of renal 69. Barr SC. Canine Chagas’ disease (American trypanosomia-
and hepatic functions in dogs naturally infected by visceral leish- sis) in North America. Vet Clin North Am Small Anim Pract.
maniasis submitted to treatment with meglumine antimoniate. Res 2009;39:1055-1064:v-vi.
Vet Sci. 2007;83:105-108.
54. Bianciardi P, Brovida C, Valente M, et al. Administration of milt-
efosine and meglumine antimoniate in healthy dogs: clinicopatho-
logical evaluation of the impact on the kidneys. Toxicol Pathol.
2009;37:770-775.
SECTION 3
Basic Principles for Infection Control
Jane E. Sykes  and  J. Scott Weese

CHAPTER 11

Infection Control Programs for Dogs and Cats


Jane E. Sykes and J. Scott Weese

w KEY POINTS
• A  hospital infection control program consists of infectious to educate staff about specific transmission precautions for
disease control personnel, a written protocol, training, and infectious diseases seen within a practice.
documentation. The aim of such a program is to reduce the • This chapter describes the infection control program and
incidence of hospital-acquired infections among patients, staff, important components of an infection control protocol, includ-
and visitors to a small animal hospital. ing the advantages and disadvantages of various methods of
• The infection control program describes the requirement for sterilization, disinfection, and antisepsis used in small animal
practices that optimize hygiene such as hand washing, the use hospitals.
of protective clothing, cleaning and disinfection, and appropri- • Information in this chapter also has relevance to infection con-
ate disposal of infectious agents. The protocol can also be used trol in shelters and in breeding and boarding facilities.

INTRODUCTION and surveillance efforts. In large hospitals, formation of an


infectious disease control committee may be required. In this
The primary role of an infection control program is to reduce the situation, the infectious disease control committee could con-
incidence of hospital-acquired infections (HAIs) by patients, staff, sist of an internist or criticalist with an interest or training in
and visitors to a small animal hospital. Infection control programs infectious diseases, a veterinary clinical microbiologist, a nurs-
in veterinary hospitals have largely evolved from evidence and ing supervisor, a safety officer, the hospital administrator, and
protocols from the human health care setting, together with our the hospital director. There should be adequate personnel and
understanding of transmission pathways for veterinary patho- communication so that proper coverage is maintained when
gens. In recent years, several factors have led to an increased rate one individual is absent or unavailable. All personnel who
of adoption of infection control programs by veterinary hospitals, work in a veterinary hospital, as well as visitors, should be
such as an increased prevalence of multidrug-resistant bacterial familiar with the infectious disease control personnel as well as
infections among small animal patients, the appearance of more procedures and policies listed in the infection control protocol.
studies that support the common occurrence of transmission of Documentation of training should be maintained for all hospi-
hospital-associated and zoonotic pathogens in the veterinary set- tal personnel.
ting, the requirement for an infection control program in veteri-
nary teaching hospitals for accreditation purposes, and increased The Hospital Infection Control Protocol
scrutiny of hospital-associated and zoonotic infections to protect
hospitals against litigation. The objective of a hospital infection control protocol is to pro-
vide a standard procedure for the control of infectious diseases
Infection Control Programs in the hospital, in order to minimize animal-to-animal, animal-
to-human, and human-to-animal transmission of pathogens.
Every small animal hospital should have an infection control Adherence to the infection control protocol can also reduce
program. At a minimum, this should consist of an infectious transmission of infectious agents between personnel through
disease control officer, a written infection control protocol, increased hand washing and reduction of fomite contamination.
regular training of staff, and documents that record all training The infection control protocol is a legal document and should

105
106 SECTION 3  Basic Principles for Infection Control

be regularly updated by a designated hospital infection control


officer or committee. Each hospital should develop a protocol BOX 11-1
that is tailored to address specific practice requirements and the
hospital design, purpose, and equipment used. Additional spe-
Guidelines for Hand Washing and the Wearing
cial precautions may need to be described for hospitals that see of Disposable Gloves
avian and exotic pet animal species, and in geographic locations
where serious zoonotic diseases such as rabies and plague are Situations That Require Hand Washing
endemic (see Chapters 13 and 55). Immediately before handling a patient
The infection control protocol details practices that optimize Immediately after handling a patient
hygiene such as hand washing, the use of protective clothing, Immediately before and after procedures that involve
cleaning and disinfection, and appropriate disposal of infectious nonintact skin
agents. Specific infectious disease control procedures to be fol- Before and after gloves are worn for procedures
lowed in different areas of the hospital (radiology, surgery, the After blood, body fluids, secretions, excretions, or con-
intensive care unit, isolation, wards) can be included, as well taminated items are touched
as policies on antimicrobial use. The protocol can also be used After cages are cleaned
to educate staff about routes of transmission, the potential for Before and after eating, smoking, or leaving the hospital
zoonotic transmission, specific transmission precautions for After going to the restroom
infectious diseases that are seen within the practice, and immu-
nization requirements, such as those for rabies (see Chapter 13). Situations When Disposable Gloves Should
Be Worn
Standard Precautions Contact with broken skin and bodily fluids (blood, urine,
and feces)
All animals that enter a veterinary hospital are potential carri- Handling of disinfectants
ers of pathogens that may be spread to other animals or people. Handling of animals with suspected or known infectious
Animals may be colonized with multidrug-resistant bacteria, diseases
often without showing evidence of disease due to these organ- Handling of all animals for immunocompromised people
isms. Pet owners may carry these organisms on their hands and
clothing. Standard precautions such as hand hygiene and the
wearing of routine protective clothing can minimize the spread
of these bacteria around the hospital and to immunocompro- has dried. Use of soap and water, rather than hand sanitizer, is
mised animals. recommended when there is gross contamination with organic
matter, or when exposure to alcohol-resistant pathogens such
Hand Hygiene as Clostridium spp. spores or parvovirus might have occurred.
Frequent and proper hand and wrist washing to remove tran- However, the availability of hand sanitizers improves compli-
sient flora on the hands has been proven as the most important ance in busy hospital situations, is associated with lower rates of
component for prevention of the spread of infectious diseases dermatitis than medicated soaps, and is recommended for rou-
in human hospitals.1 Signs that outline proper hand-washing tine hand sanitation in human health care settings by the Cen-
technique, including the use of paper towels to turn off the fau- ters for Disease Control and Prevention (CDC) and the World
cet, posted adjacent to basins around the hospital can improve Health Organization (WHO).3,4
compliance among staff. Online videos that demonstrate proper The use of gloves prevents contamination of the hands with
hand-washing technique are available for educational purposes.2 microorganisms, prevents exposure to bloodborne pathogens,
Guidelines for hand washing are shown in Box 11-1. Antibacte- and reduces the risk of transmission of microorganisms from
rial soap should be used, and all surfaces of the hands should be personnel to animals. However, gloves are not a substitute for
rubbed together, which should include the backs of the hands, proper hand hygiene. Guidelines for wearing disposable gloves
between the fingers, and under the fingernails, for a total hand- are shown in Box 11-1. Gloves should be promptly removed
washing time of at least 20 seconds. In order to prevent chapped after use, before other surfaces are touched, and hands should
skin, which can harbor bacteria, water used for hand washing then be washed. If a glove is torn or punctured, it should be
should not be too hot, and hand lotion should be applied regu- removed and replaced as soon as possible.
larly. Behaviors such as keeping fingernails short, avoidance of
artificial and/or polished fingernails or hand jewelry, or wearing Hospital Attire
jewelry on a chain around the neck instead of on the hand can Nonsurgical Areas
be encouraged. Meticulous hand hygiene is particularly impor- Staff should be encouraged to wear dedicated hospital attire
tant for personnel who work frequently with immunocompro- that is not worn elsewhere, so that hospital pathogens are not
mised animals, such as emergency and critical care personnel. transported to and from locations outside the hospital. At the
The use of touch-free taps and paper towel dispensers can also minimum, protective clothing such as clean laboratory coats or
reduce transmission of bacteria during hand washing. hospital scrubs and closed-toe shoes must be worn in nonsurgi-
Alcohol-based hand sanitizers are a more convenient form of cal areas. Sleeves must be short enough or rolled up to expose
sanitization. These can be provided in multiple locations around the wrists, and laboratory coats changed whenever gross soiling
a hospital, or travel-sized bottles can be carried in a coat pocket. occurs. In the absence of gross soiling, coats should be changed
At least one to two full pumps or a 3-cm diameter pool of the daily. If neckties are worn, they must be secured in place by an
product should be dispensed onto one palm. All surfaces of the outer layer of clothing or a tie pin so that they cannot be con-
hands and wrists should be rubbed with the product until it taminated as a result of contact with patients or environmental
CHAPTER 11  Infection Control Programs for Dogs and Cats 107

TABLE 11-1
Transmission Precautions for Selected Infectious Diseases of Dogs and Cats
Infectious agent Specific precautions
Airborne Mycobacterium tuberculosis Isolation, preferably in negative pressure facility
Yersinia pestis Properly fitted N95 respirator masks
Francisella tularensis Contact precautions
Droplet Canine and feline transmissible respiratory disease pathogens Isolation
(e.g., canine distemper virus, canine respiratory coronavirus, Space animals at least 4 feet apart
feline herpesvirus-1, feline calicivirus) Contact precautions
Contact Multidrug-resistant bacteria Warning signage
Dermatophytes Gown, gloves, dedicated equipment
Leptospira spp. Isolation for select pathogens (see Box 11-2)
Salmonella spp. Limitation of movement
Parvoviruses Standard hand hygiene precautions
Proper cleaning, disinfection, and disposal of
medical waste

surfaces and act as fomites, as has been shown to occur in and warning signage on cages and medical records. Should bites
human hospital environments.5 Long hair must be tied back so or scratches occur, they should be vigorously flushed and imme-
that it does not drape on animals and hospital surfaces. Face diately washed with water and chlorhexidine or a dilute iodo-
shields and a clean gown should be worn during procedures phor solution, and the bite reported to appropriate officials as
that are likely to generate splashes or sprays of blood and body necessary. Deep wounds could be irrigated with pressure using
fluids. Gowns must be made of impervious material and tied on a syringe without an attached needle. If a bite occurs, medi-
securely and correctly. Soiled gowns must be removed as soon cal attention should be sought as soon as possible. All bites or
as they are no longer required and face shields cleaned. scratches must be documented and consideration be given as to
why the injury occurred, so that procedures or training to pre-
Surgical Areas vent future injuries can be implemented if necessary.
Dedicated operating room attire should be worn in surgical Consumption of food and drink should be limited to parts of
areas and should be changed after gross soilage and when leav- a hospital where patient care and the handling of biologic speci-
ing the operating room for the day. Caps and masks should mens and medications do not occur. Food and beverages should
be worn and hands thoroughly washed on entry to the operat- not be left out open on benches for long periods. Microwaves
ing room. There is no evidence that dedicated footwear or foot used for animal care purposes should not be used to heat food
protection should be worn for prevention of surgical site infec- intended for people.
tions in human patients, and major human guidelines do not
advocate that footwear be addressed.6 Protective wear should Transmission-Based Precautions
be removed whenever staff members leave the operating room. Transmission-based precautions are instituted for selected
Traffic in and out of the operating room should be limited to the patients that are confirmed to be or suspected to be infected
minimum required for patient care. or colonized with important transmissible pathogens. Trans-
mission-based precautions are used in combination with stan-
General Animal Handling Precautions dard precautions. In the human hospital setting, three types
All animals seen at a veterinary hospital should undergo a his- of transmission-based precautions have been developed—air-
tory and physical examination by a veterinarian to determine borne, droplet, and contact precautions (Table 11-1).8 Airborne
the likelihood and nature of any transmissible infections that precautions are used to prevent the transmission of diseases by
might be present. Ideally, client beds, blankets, collars, and droplet nuclei (particles <5 µm). Transmission by droplet nuclei
leashes should not be brought into the hospital, where they occurs with diseases such as measles, varicella, and pulmonary
could become contaminated. Animals should always be placed tuberculosis. The precautions for human patients involve isola-
in cages that have been cleaned and disinfected appropriately. tion in a single-bed, negative-pressure room and the wearing
Disposable thermometer sleeves should always be used on ther- of high-density respirator (N95) masks. These resemble surgi-
mometers. Equipment should not be shared between animals cal masks but filter 1-µm particles with an efficiency of at least
unless it has been cleaned and disinfected. Diets that contain 95% and must be properly fitted. Airborne contact precautions
raw meat and bones should not be fed or stored in the hospital, are rarely necessary in hospitals that treat only dogs and cats,
because they commonly contain and can potentially transmit but could be considered when animals suspected to have pneu-
foodborne gastrointestinal pathogens.7 The handling of sick monic plague, tularemia, or Mycobacterium tuberculosis infec-
animals should be minimized, unless required for patient care. tion. Droplet precautions are used to prevent transmission by
Because some infections can be transmitted through bites large-particle aerosols and do not require a negative-pressure
and scratches, staff should be educated on bite and scratch room. Droplet precautions apply to dogs and cats with trans-
avoidance, such as the use of restraint devices, protective gloves, missible respiratory disease. Contact precautions are indicated
108 SECTION 3  Basic Principles for Infection Control

for animals with infections that can be transmitted by direct visible and/or accessible and may not provide access to an oxy-
contact with the patient or through fomite contact. gen source or be amenable to intensive monitoring and care. For
If it is known in advance that an animal with a suspected some animals (especially puppies and kittens) suspected to have a
or known transmissible disease is to be seen at the hospital, transmissible disease, housing in a general ward or ICU area with
arrangements should be made to have the pet owner take the as much physical and procedural separation as possible, and with
animal from the parking area directly to an examination room, strict infection control practices, may be acceptable (if perhaps
so that the animal does not contaminate the waiting area. After not optimal). Once a diagnosis is confirmed, the animal should
the animal has been examined, appropriate signage should be be moved immediately to isolation whenever possible. Patients
placed on the door of the examination room to prevent use until chosen for strict isolation vary based on the specific situation
it has been properly cleaned and disinfected. and facilities available, but suggestions are provided in Box 11-2.
If hospitalization is required, animals with known or suspected Only the individuals directly involved in the care of the patient
transmissible disease must be admitted either to an isolation ward should enter isolation. Pet owners should not be allowed into
or to regular hospital cages with handling precautions, based on the isolation ward. No equipment used outside isolation (pens,
the pathogen suspected. Immediately after hospitalization, the thermometers, stethoscopes, cell phones) should be brought into
name of the suspected or known pathogen should be posted on isolation. Laboratory coats should be removed, and personnel
the cage or run, together with a handling precautions notice. Ani- must put on protective wear such as a disposable gown, gloves,
mals that require contact precautions should be placed in cages and booties when entering the isolation ward. Face protection
away from other animals in the ward and should not be moved may also be required, depending on the situation. A notice that
from one cage to another, unless there is a medical need. Animals outlines the required precautions should be posted on the door
with transmissible respiratory disease should be placed in isola- of isolation. Protective clothing should be removed before leav-
tion and separated both horizontally and vertically from other ing isolation, and hands should be washed. Once a patient has
patients by at least 4 feet. Gloves and a gown should be put on been discharged, the room should be properly disinfected.
before the patient is handled, and then removed and disposed of
immediately afterward. Hands should be washed after the gloves Handling of Potentially Infectious Materials
are removed. Additional items of personal protective equip- and Waste
ment (masks, gowns, gloves, booties) may be required in some The risk of human infection from patient blood and body fluids
circumstances. Soiled linen and equipment should be handled, in small animal hospitals is clearly lower than that in human
transported, and processed in a manner that prevents skin and hospitals. However, a number of zoonotic pathogens have the
mucous membrane exposures and contamination of clothing and potential to be spread from dogs or cats to humans through
that avoids transfer of microorganisms to other patients and envi- contact with medical waste, and a number of emerging zoonotic
ronments. All equipment that contacts the patient (scales, exami- infectious agents are bloodborne. Examples include Bartonella
nation tables, stethoscope heads, floor) should be cleaned and spp., Anaplasma phagocytophilum, Brucella spp., and hemo-
disinfected immediately after use. Medications and fluids from plasmas. Proper handling of blood and body fluids also has the
these patients should not be returned to the hospital pharmacy. potential to protect staff against as-yet-unrecognized zoonotic
When possible, personnel who handle these animals should not pathogens. Improper disposal of medical waste in veterinary
work with other immunosuppressed animals in the hospital, or hospitals risks injuring others who handle the waste and has the
they should work with the infectious disease cases last. If animals potential to result in serious penalties or fines.
with contact precautions must be moved within the hospital, per- Potentially contaminated waste should be disposed of in an
sonnel should ensure that precautions are maintained throughout approved plastic bag in a container labeled on all exposed sides
and that equipment and environmental surfaces that come into
contact with the patient are properly disinfected and/or disposed
of. The cleaning and bandaging of wounds infected with multi-
drug-resistant bacteria should be conducted in low-traffic areas BOX 11-2
that can be properly cleaned and disinfected.
Examples of Infectious Agents of Dogs and Cats
The owners of dogs and cats that have transmissible diseases
should be provided with general information regarding the risk
for Which Strict Isolation Is Indicated
of disease transmission to in-contact animals and people that
includes the mode of transmission, duration of organism shed- Salmonella spp.
ding, and if there are special implications for young children or Francisella tularensis
other immunosuppressed individuals. If dogs and cats are diag- Yersinia pestis
nosed with a zoonotic disease, the owners should be notified Mycobacterium tuberculosis or Mycobacterium bovis
without delay. The owners should be told to see their physicians Microsporum canis
if they become unwell or, in some circumstances, immediately, Rabies virus
and to advise a physician of the potential exposure. Enteric viruses, such as parvoviruses
Canine transmissible respiratory disease pathogens
Isolation (Bordetella, canine distemper virus, influenza viruses,
In contrast to human hospitals where patients can be more read- canine respiratory coronavirus, canine adenovirus,
ily isolated in single-bed rooms or cubicles, isolation of veteri- canine parainfluenza virus)
nary patients can be more difficult because of the close proximity Feline upper respiratory tract disease pathogens (feline
of one animal to another. Floor contamination with secretions herpesvirus-1, feline calicivirus, influenza virus,
and excretions can also occur more readily. Isolation rooms are Chlamydia)
available in many veterinary hospitals, but they may be poorly
CHAPTER 11  Infection Control Programs for Dogs and Cats 109

with “biohazardous waste.” Blood-soaked materials, infected masses or peripheral lymph nodes are aspirated or venipuncture
materials, and empty fluid bags should all be placed in biohaz- is performed. Contaminated sharps (slides, scalpel blades, broken
ardous waste containers. Care should be taken not to contami- glass, needles with attached syringes) must immediately be placed
nate the outside of the container during disposal. The lid of the into an approved puncture-resistant sharps disposal biohazard
biohazardous waste container must close properly. Blood and container. Personnel who perform necropsy examinations should
body fluids should be inactivated with an appropriate disinfec- take care to use sharp knives in the proper manner and to avoid
tant (e.g., bleach or accelerated hydrogen peroxide) and allowed rushed situations. If a contaminated sharps injury occurs, medical
to stand for 10 minutes before disposal. Disposal regulations attention should be sought immediately if necessary.
may vary depending on local laws, but liquid waste should not
be disposed of into storm drains. Hospital Cleaning, Disinfection, and Sterilization
Specimens for laboratory testing that are collected from a Definitions
patient with suspected zoonotic infectious diseases should be Sterilization refers to complete elimination of all microbes,
placed in an outer plastic bag, taking care not to contaminate including bacterial spores, and is accomplished in hospital set-
the outside of the plastic bag, and labeled with an appropri- tings using processes such as pressurized steam, dry heat, ethyl-
ate warning label. Fecal material should be picked up using a ene oxide gas, or liquid chemicals.
tongue depressor while wearing gloves, and placed in a sealed Disinfection is the process that eliminates many or all
plastic cup and clearly labeled. Urine specimens should be sub- microbes from inanimate objects, but not bacterial spores. Fac-
mitted in a sealed container. tors that influence the efficacy of disinfection include the type of
microorganism present, their number, the amount and type of
Sharps Handling organic matter present, the presence of biofilms, and the porosity
Sharps handling practices receive considerable attention in human of the surface to be disinfected. Some disinfectants kill spores at
medicine because of the risk of transmission of various blood- high concentrations and with prolonged exposure times. These
borne pathogens. Although the risks are less in veterinary medi- are known as chemical sterilants. At low concentrations and
cine, significant injury or illness can follow sharps injuries, such short contact times, chemical sterilants inactivate all microbes
as transmission of infectious agents from the patient, allergic or except large numbers of bacterial spores and are known as
inflammatory reactions from exposure to medication, and inocu- high-level disinfectants. Low-level disinfectants inactivate most
lation of opportunistic pathogens from the injured person’s own vegetative bacteria, some fungi, and enveloped viruses, but
skin microflora. Care should be taken to prevent injuries when not bacterial spores. Intermediate-level disinfectants inactivate
needles, scalpels, and other sharp instruments are used, cleaned, mycobacteria, vegetative bacteria, most viruses, and most fungi
or disposed of. Used needles should never be recapped, and they (Tables 11-2 and 11-3).
should not be removed from the barrel of a disposable syringe. Antisepsis is the process that reduces the number of microbes
Personnel should be instructed not to walk with uncapped nee- from living tissue and skin. Disinfectants are rarely used for skin
dles and not to hold syringe or needle caps in the mouth. Used antisepsis because they can injure tissues and skin. Sanitation is
needles should not be carried in a pocket. Animals suspected of the reduction in the number of microorganisms on a surface to a
having an infectious disease that could be transmitted to humans safe level. Germicides are agents that inactivate microorganisms
through a needle-stick injury should be sedated before skin and include disinfectants, antiseptics, and sanitizers.

TABLE 11-2
Characteristics of Selected Disinfectants
Activity in the
Disinfectant ­Presence of
Category ­Organic Matter Advantages Disadvantages Precautions Comments
Alcohols: Rapidly Fast-acting Rapid evaporation Flammable Not appropriate for
Ethyl alcohol, inactivated No residue routine environmen-
isopropyl Relatively nontoxic tal dis­infection
alcohol Primarily used as
­antiseptics
Aldehydes: Good Broad spectrum Highly toxic Irritant Used as an aqueous
Formaldehyde, Relatively noncor- Carcinogenic solution or as a gas
glutaraldehyde rosive Requires ventilation (fumigation)
Alkalis: Unpleasant odor Do not mix with Not recommended for
Ammonia Irritating bleach. general use
Biguanides: Rapidly Nontoxic Incompatible with Not appropriate for
Chlorhexidine inactivated anionic detergents ­environmental dis­
infection
Primarily used as
­antiseptics
Continued
110 SECTION 3  Basic Principles for Infection Control

TABLE 11-2
Characteristics of Selected Disinfectants—cont’d

Activity in the
Disinfectant ­Presence of
Category ­Organic Matter Advantages Disadvantages Precautions Comments
Halogens: Rapidly Broad spectrum, Inactivated by Corrosive Used to disinfect clean
Hypochlorites inactivated sporicidal cationic soaps/ Irritant environmental
(bleach) Inexpensive detergents and May produce toxic ­surfaces
Can be used on sunlight. gas when mixed Only commonly
food preparation Frequent applica- with other chemi- available sporicidal
surfaces tion required. cals disinfectant
Oxidizing agents Good Broad spectrum Breakdown with Corrosive Excellent choice for
Environmentally time environmental
friendly ­disinfection
Phenolics Good Broad spectrum Toxic to cats Irritant Some residual activity
Noncorrosive Unpleasant odor after drying
Stable in storage Incompatible with
cationic or non-
ionic detergents
Quaternary Moderate Stable in storage Incompatible with Commonly used
­ammonium Nonirritating to skin anionic deter- ­primary environmen-
­compounds Low toxicity gents tal disinfectant
(QUATs) Can be used on food Some residual activity
preparation surfaces after drying
Effective at high tem-
peratures and pH

Source: Modified from the Canadian Committee on Antibiotic Resistance. Infection Prevention and Control Best Practices for Small Animal Veteri-
nary Clinics, 2008; http://www.wormsandgermsblog.com/2008/04/promo/services/infection-prevention-and-control-best-practices-for-small-animal-
veterinary-clinics/. Last accessed May 15, 2012.

TABLE 11-3
Antimicrobial Spectrum of Selected Disinfectants
Halogens: Quaternary
Alkalis: Biguanides: H­ ypochlorite Oxidizing Ammonium
Agent Alcohols Aldehydes Ammonia Chlorhexidine (Bleach) Agents Phenolics Compounds
Mycoplasmas ++ ++ ++ ++ ++ ++ ++ +
Gram-positive bacteria ++ ++ + ++ ++ ++ ++ ++
Gram-negative bacteria ++ ++ + + ++ ++ ++ +
Pseudomonads ++ ++ + ± ++ ++ ++ ±
Enveloped viruses + ++ + ++ ++ ++ ++ +
Non-enveloped viruses – + ± – ++ + ±* –
Fungal spores ± + + ± + ± + ±
Mycobacteria + ++ + – + ± ++ –
Bacterial spores – + ± – ++ + – –

++, Highly effective; +, Effective; ±, Limited activity; –, No activity.


*In general, phenols are not active against non-enveloped viruses but they do have some activity against rotaviruses. Activity against parvoviruses
has not been documented.
Examples of microorganisms from each category:
Mycoplasmas: Mycoplasma cynos, Mycoplasma felis; Gram-positive bacteria: Staphylococcus spp., Streptococcus spp.; Gram-negative bacteria: Bor-
detella bronchiseptica, Salmonella spp.; Pseudomonads: Pseudomonas aeruginosa; Enveloped viruses: influenza virus, herpesvirus; Non-enveloped
viruses: feline panleukopenia virus, canine parvovirus, feline calicivirus; Fungal spores: Aspergillus spp.; Acid-fast bacteria: Mycobacterium fortui-
tum; Bacterial spores: Clostridium difficile, Clostridium perfringens.
Source: Modified from the Canadian Committee on Antibiotic Resistance. Infection Prevention and Control Best Practices for Small Animal Veteri-
nary Clinics, 2008; http://www.wormsandgermsblog.com/2008/04/promo/services/infection-prevention-and-control-best-practices-for-small-animal-
veterinary-clinics/. Last accessed May 15, 2012.
CHAPTER 11  Infection Control Programs for Dogs and Cats 111

TABLE 11-4
Recommended Conditions for Heat Sterilization
Conditions Time
Gravity displacement steam 121°C (250°F), 106 kPa (15 lb/in2)* 30 min wrapped items, 20 min unwrapped
sterilization 132°C (270°F), 30 lb/in2 15 min wrapped items
High-speed prevacuum steam 132°C (270°F), 30 lb/in2 4 min
sterilization
Dry heat sterilization 160°C (320°F) 2 hours at temperature, 3 to 3.5 hours with cooling
170°C (340°F) 1 hour at temperature, 2 to 2.5 hours with cooling

*Pressure settings may vary between incubators. When possible, follow manufacturer’s recommendations.

Cleaning involves the removal of all visible organic and prevacuum sterilizers rapidly pump air out of the sterilizer
inorganic material from objects and surfaces through the use before steam is admitted. This leads to rapid penetration of
of manual or mechanical processes and detergent or enzymatic steam into all surfaces. As a result, cycle times can be reduced
solutions. to less than 15 minutes. Drying times are also reduced with
According to the Spaulding method of classification, items prevacuum sterilizers.
to be sterilized or disinfected can be grouped into critical, semi- Sterilizers should be located away from potential sources of
critical, and noncritical items.9 Critical items enter tissue or the contamination, such as sinks, trash disposal, or high-traffic areas.
vascular system, or devices through which blood flows. Critical Before steam sterilization is performed, instruments should be
items require sterilization before they can be used. Semicritical cleaned thoroughly to remove organic and inorganic material,
items are items that contact mucous membranes or nonintact and then dried. All jointed items should be opened or unlocked,
skin and include endoscopes and balloon dilation catheters. and items should not be crowded in the autoclave, so that steam
These generally require high-level disinfection. Noncritical can circulate freely. At least 3 inches should be left between the
items are items that contact intact skin. Noncritical items gener- autoclave wall and items to be sterilized. The manufacturer’s
ally require low-level or intermediate-level disinfection and con- instructions for autoclave operation should be followed.
tact times of 1 to 10 minutes.9 The efficacy of autoclaving must be tested for every auto-
All personnel should be educated regarding the standard claved item, with additional quality control performed on
hospital germicides used for disinfection and antisepsis, how a periodic basis. Autoclave indicator tape is routinely used;
they should be diluted and applied, and the hazards associated although this only indicates conditions on the outside of the
with their use. Gloves and eye protection should be worn when package. Steam indicator strips should be included in each
disinfectant solutions are handled or mixed. In large hospitals, surgical pack and evaluated by the person opening the pack.
posters on hospital walls can be used to guide selection of appro- Biological indicators provide a more definitive assessment of
priate germicides for different situations. Disinfectant solution autoclave efficacy and should be used periodically (e.g., weekly)
should be readily accessible throughout the hospital. Because and the results documented. These consist of a standardized
mops can spread infection, cotton mops should be laundered population of bacterial spores, usually on a filter paper strip
daily, or mops with detachable microfiber heads should be used. or contained within a vial. The strip is then sent to the micro-
Microfiber heads absorb a large amount of water and are not biology laboratory for culture, or cultured in-house, to ensure
returned to the mop bucket after use. They can be laundered that the spores have been completely inactivated by the steril-
and reused the following day. ization process. Any indicator failure should result in immedi-
ate inspection of the autoclave. Sterilization indicators should
Methods of Sterilization never be used as a substitute for proper autoclave operation
Steam sterilization.  Steam sterilization involves the use and careful preparation, packing, and loading of equipment to
of saturated steam under pressure in an autoclave to achieve be sterilized.
sterilization. This is the most effective form of steriliza- After steam sterilization, instruments should be allowed to
tion, is nontoxic and inexpensive, and as a result is the most dry before they are removed, which typically takes an additional
widely used as well. The use of steam under pressure allows 30 minutes. Items should then be stored in a location and man-
lower temperatures to be used for shorter periods of time ner that prevents further contamination.
when compared with dry heat sterilization (Table 11-4). The Flash sterilization.  Flash sterilization refers to the rapid ster-
most common temperatures used for steam sterilization are ilization of unwrapped instruments and is usually performed as
121°C (250°F) and 132°C (270°F). Cycle times vary depend- an emergency procedure in an operating room setting when time
ing on the autoclave used and whether items are wrapped or is insufficient to perform the preferred sterilization of wrapped
unwrapped, but are generally less than 30 minutes (see Table items. In general it is performed for 3 minutes at 270°F and 27 to
11-4). There are two basic types of autoclaves: gravity dis- 28 lb/in2. Each instrument must be carefully protected to ensure
placement autoclaves, and high-speed prevacuum sterilizers. it does not become recontaminated during transport back to the
Gravity displacement autoclaves admit steam at the top or operating room, usually in a “flash pan.” Flash sterilization has
sides of the autoclave, which displaces air through a drain occasionally been associated with intraoperative infections and
vent at the bottom of the chamber (Figure 11-1). High-speed should not be used for routine disinfection purposes.
112 SECTION 3  Basic Principles for Infection Control

FIGURE 11-1  Gravity displacement sterilization. Steam is admitted at the top or sides of the autoclave, which displaces air through a drain vent at the bottom of the chamber.

Gas sterilization.  Gas sterilization is used for sterilization contact times and at the right concentration. Solutions may lose
of heat- and moisture-sensitive instruments but can have sig- efficacy, become diluted, or become contaminated with bacte-
nificant toxicity. Gases that can be used for sterilization include ria over time. They should be replaced frequently according to
formaldehyde, ethylene oxide (ETO), hydrogen peroxide vapor, the manufacturer’s recommendations, and they should never be
and ozone gases. The most commonly used gas in veterinary used in procedures that involve sterile body sites unless there is
medicine, ETO, has strong alkylating properties and causes pro- no other sterilizing option.
tein coagulation, enzyme inactivation, and damage to nucleic
acid. When compared with heat sterilization, ETO sterilization Disinfectants
takes longer (24 hours or more including the time required to Glutaraldehyde.  Glutaraldehyde is a saturated dialdehyde.
allow ETO to diffuse out of packages at the end of sterilization) Glutaraldehyde solutions are relatively inexpensive, are non-
and is more expensive. In addition, it only achieves surface ster- corrosive, and can be used to disinfect rubber, plastics, and
ilization and requires sophisticated equipment and trained staff. endoscopic equipment. Aqueous solutions require activation
The gas is extremely flammable, irritates the eyes and mucous by alkalinization to a pH of 7.5 to 8.5 for sporicidal activity
membranes, is mutagenic and carcinogenic, and has a mislead- to occur. When alkalinized glutaraldehyde is used at a concen-
ingly pleasant smell. tration of 2.4% for adequate periods of time, either chemical
Irradiation.  Gamma irradiation uses a cobalt-60 radiation sterilization or high-level disinfection occurs, depending on the
source to destroy microorganisms through generation of high- contact time (e.g., Cidex). Contact times of at least 20 minutes
energy photons. Health care product manufacturers use gamma (at or above 20°C) are effective for high-level disinfection. Ster-
irradiation to sterilize disposable medical supplies, such as cath- ilization requires a 10-hour contact time and higher concentra-
eters, gloves, syringes, and pharmaceuticals. tions of glutaraldehyde (e.g., Cidex Plus, which contains 3.4%
Chemical sterilants.  Chemical sterilants are used when heat glutaraldehyde), or formulations that combine glutaraldehyde
or ETO gas sterilization is not available or would otherwise with another disinfectant.
damage instruments, such as endoscopes or laparoscopes. Dis- Once activated, 2.4% glutaraldehyde solution retains activity
infectants that act as sterilants when they are used at high con- for 14 days, provided inadvertent dilution does not occur. The
centrations and for adequate contact periods include certain solution is active in the presence of 2% organic matter. Inad-
solutions that contain glutaraldehyde (e.g., >2.4% glutaral- vertent dilution can occur when endoscopes that contain fluid
dehyde solutions, 0.95% glutaraldehyde with 1.64% phenol/ within their channels are immersed in the solution. Test strips
phenate), 0.55% ortho-phthalaldehyde (OPA), 7.5% hydrogen are available from the manufacturer to monitor the activity of
peroxide, or greater than or equal to 0.2% peracetic acid (see the solution, but should not be used to extend the solution’s
section on Disinfectants following). Chemical sterilants must be expiration date. They indicate inactivity when the concentra-
rinsed repeatedly with sterile water and dried once sterilization tion drops below 1.5%, the minimum concentration required
is complete. Chemical sterilants must be used with the proper for activity.
CHAPTER 11  Infection Control Programs for Dogs and Cats 113

Glutaraldehyde irritates mucous membranes of the respi- 7 days. The powder is corrosive and can cause serious skin and
ratory and gastrointestinal tracts, and so endoscopes must be ocular burns, but the solution is nonirritating and less corrosive
rinsed properly after disinfection. It can also cause allergic than bleach. The solution stains fabric and may damage sur-
contact dermatitis, but it is not mutagenic or carcinogenic. faces, particularly metal, over time if rinsing is not performed.
The wearing of nitrile rubber or butyl rubber gloves is recom- Sodium Hypochlorite (Bleach).  Household bleach, which con-
mended. Because of its relative expense and toxicity, it is not tains 5.25% to 6.15% sodium hypochlorite, is widely available,
used to disinfect noncritical surfaces. inexpensive, and active in the presence of hard water. When
ortho-Phthalaldehyde.  OPA (e.g., Cidex OPA) is favored over used at a 1:10 dilution for a 10-minute contact time, household
glutaraldehyde for high-level disinfection in the United States, bleach is sporicidal but is irritating and can be highly corro-
because it does not require activation, is stable over a wide pH sive to metal surfaces. This 1:10 dilution is used to control out-
range, does not cause irritation of mucous membranes, and it breaks of clostridial diarrhea. For most hospital situations, 1:30
has a barely perceptible odor. Its activity is greater than that of to 1:50 dilutions of household bleach provide more than 1000
glutaraldehyde, and high-level disinfection is achieved with a ppm available chlorine and are effective for intermediate-level
contact time of 12 minutes at or above 20°C.10,11 The primary disinfection. Noncritical surfaces can be disinfected with 1:500
disadvantage of OPA is that it stains tissues and mucous mem- dilutions of household bleach (>100 ppm available chlorine),
branes gray. Protective equipment must be worn when handling with contact times of at least 1 minute. Sodium hypochlorite
the solution, and it must be rinsed thoroughly from items after is inactivated by organic matter, so cleaning is required before
treatment. Irritation can occur with eye contact. OPA is also disinfection is performed. Solutions lose 50% of their activity
more expensive than glutaraldehyde.12 Solutions can be reused over a 1-month period unless they are stored in closed brown
for a maximum of 14 days. bottles. Other disadvantages of bleach solutions are bleaching
Hydrogen peroxide.  Hydrogen peroxide (H2O2) is a potent of colored fabric and the release of toxic chlorine gas when
oxidizer. Hydrogen peroxide solutions are widely available, are they are combined with other disinfectants such as quaternary
inexpensive, and can enhance removal of organic matter. When ammonium compounds (QUATs).
used at concentrations of 7.5% for contact periods of at least Quaternary Ammonium Compounds.  QUATs are often used
6 hours, hydrogen peroxide is a chemical sterilant (e.g., Spo- as low-level disinfectants for noncritical surfaces in health care
rox). High-level disinfection can be achieved with contact times facilities. Although they are generally fungicidal, bactericidal,
of 12 to 30 minutes. Unfortunately 7.5% hydrogen peroxide and virucidal for enveloped viruses, some bacteria resist and
causes discoloration and functional changes within endoscopes even grow within QUATs, and such contamination has resulted
and so is not suitable for endoscope reprocessing. Although in HAIs.17 Contact times vary by product so manufacturer rec-
nonirritating to mucous membranes, serious ocular damage can ommendations should be followed; however, 10-minute contact
occur with eye contact. times are often used. Advantages of QUATs include their low
Accelerated hydrogen peroxide (AHP) is a patented hydrogen cost, high stability, and low toxicity. QUATs are inactivated by
peroxide solution that contains surfactants, an acid, and hydro- hard water, organic materials, soaps, and detergents.
gen peroxide. Use of a 4.5% AHP gel with contact times of 10 Phenolics.  Phenolics are one of the oldest known disinfec-
minutes can inactivate bacterial spores.13 Even 0.5% solutions tant classes. Phenol derivatives that are available for hospital
have some sporicidal activity and can inactivate non-enveloped disinfection include ortho-phenylphenol and ortho-benzyl-para-
viruses such as canine parvovirus.14,15 AHP has gained popular- chlorophenol (e.g., Qualitrol, Vetnex). Effective contact times
ity as a disinfectant among health care institutions because it is vary with the product. They are active against enveloped viruses
odorless, does not generate volatile gas, is nonirritating, and is and bacteria, but they are not sporicidal and have limited activity
noncorrosive at dilutions used in health care settings. against fungi and non-enveloped viruses. Phenol derivatives are
Peracetic acid.  Peracetic acid belongs to the peroxygen fam- active in the presence of organic matter and hard water, are stable,
ily of compounds. When used at 50°C to 56°C in a specific per- and are noncorrosive. They irritate skin and mucous membranes,
acetic acid reprocessing system (Steris System 1, Steris), 0.2% and have the potential to be highly toxic if ingested by cats.
solutions achieve sterilization in very short time periods (30 to Because of these limitations and the availability of other effective
45 minutes). Peracetic acid is active in the presence of organic disinfectants, phenolics are rarely used in veterinary hospitals.
matter and may actually enhance its removal. Nevertheless
endoscopes must still be thoroughly cleaned before sterilization Antiseptics
to avoid fixation of blood onto the instrument. After steriliza- Alcohol.  Ethyl alcohol and isopropyl alcohol are bactericidal,
tion, the processor rinses the instrument thoroughly. Peracetic virucidal (for enveloped viruses), and variably fungicidal. The
acid is stable but can be corrosive and causes discoloration of optimum bactericidal concentration is 60% to 90% in water
endoscopes over time. It is more expensive than other chemi- (volume/volume). They do not destroy bacterial spores or pen-
cal sterilants. Peracetic acid concentrates can cause irritation to etrate proteinaceous material and so are not recommended for
mucous membranes and are corrosive to the eye and skin, but high-level disinfection. They can be used to disinfect noncriti-
0.2% solutions are generally nonirritating. cal items such as hospital fomites and have very low toxicity,
Potassium peroxymonosulfate (Trifectant, Virkon S).  Like per- so they are often included in waterless hand sanitizer products
acetic acid, potassium peroxymonosulfate is an oxidizing agent, together with emollients to prevent drying of the skin. They are
and 1% solutions are high-level disinfectants that are capable flammable and dry quickly, so it can be difficult to achieve ade-
of inactivation of non-enveloped viruses when contact times of quate contact times (≥1 minute). As a result, alcohol should not
10 minutes are used.16 Thus it is suitable for inactivation of be used for routine environmental disinfection.
canine parvovirus and feline calicivirus. Potassium peroxy- Iodophors.  Iodine solutions are primarily used as skin anti-
monosulfate retains some activity in the presence of organic mat- septics. Formulations for disinfection are also available, which
ter. Solutions are prepared from powder and remain active for contain higher concentrations of free iodine than antiseptic
114 SECTION 3  Basic Principles for Infection Control

preparations. An iodophor is a combination of iodine and a sol- responsible for cleaning and disinfection at each time point, sit-
ubilizing agent (e.g., polyvinylpyrrolidone in povidone-iodine), uation, and location in the hospital should be clearly identified.
which serves to provide a sustained-release form of iodine. A plan for the cleaning of outdoor areas that become contami-
Dilutions of iodophors are more active against microbes than nated with excretions or other biohazardous material should be
concentrated povidone-iodine, so iodophores must be diluted outlined.
correctly. Iodophors are bactericidal and virucidal. Their anti- When disinfection of cages or runs is performed, animals
bacterial activity does not persist for long periods on skin or should be removed and placed in a clean holding cage or run,
in tissues, so frequent reapplication is required. Iodophors are away from other patients. Solid waste and soiled laundry or
relatively nontoxic and nonirritating. They are stable in solu- paper should be removed and placed directly in waste or laun-
tion but are inactivated by organic material and they can stain dry containers, taking care to avoid dripping onto the floor. If
plastics and, to some extent, tissues. laundry is contaminated with potentially infectious material, it
Chlorhexidine.  Chlorhexidine is a cationic bisbiguanide that must be bagged and labeled with the contents and suspected
disrupts microbial cell membranes and precipitates cell con- infectious agent. Proper contact times should be used after
tents. It is used widely for skin antisepsis in veterinary medicine. application of disinfectant solution. Runs and cages should be
Chlorhexidine has persistent activity on the skin, is nonirritat- allowed to dry completely before a new patient is introduced.
ing, is active in the presence of body fluids, and has rapid bacteri-
cidal activity. Like iodophors, chlorhexidine has limited activity Immunocompromised People and Children
against fungi and mycobacteria. A 2% chlorhexidine solution is
preferred over 70% alcohol or povidone-iodine for skin prepa- People are considered immunocompromised if they (1) have
ration of central venous catheter sites in human patients18 and various comorbidities such as diabetes mellitus, chronic kidney
is the skin antiseptic of choice for collection of blood for blood failure, leukopenia, immune-mediated disease, HIV/AIDS, con-
cultures. Chlorhexidine may have slightly lower activity against genital immunodeficiencies, hepatic cirrhosis, cancer, or sple-
gram-negative bacteria and fungi than povidone-iodine, and nectomy; (2) are being treated with immunosuppressive drugs
both chlorhexidine and iodophors have a slower antimicrobial or chemotherapeutics; (3) are very young (5 and under) or
activity than 60% to 90% alcohol solutions. very old; or (4) are pregnant, although the immunosuppressive
effects of pregnancy are considerably lower than those of the
Principles of Cleaning and Disinfection preceding disorders.
Standard operating procedures that describe preparation and Immunocompromised staff members who work in small
application of disinfectants for all surfaces and objects used in animal hospitals should discuss any necessary work restrictions
a veterinary hospital, as well as waste management, should be and precautions in light of their specific condition with their
developed, and staff and visitors should receive education about physician or an infectious disease doctor. The risks for a dia-
the location of protocols and their use. Wards and procedure betic, for example, may be significantly different from those for
and examination rooms should remain uncluttered in order to a transplant recipient. In general, immunosuppressed individu-
facilitate effective cleaning. Because organic matter can inacti- als who work in small animal hospitals should avoid handling
vate disinfectants, all visible debris should be removed before patients with suspected or known infectious diseases. Gloves
disinfection. Disinfectants are only active when applied to clean, should always be worn when handling animals and animal
nonporous surfaces. Porous surfaces such as dirt and wood can- fluids or excreta, and strict attention to general hand hygiene
not be effectively disinfected using routine procedures. Hard and any bites, scratches, or sharps injuries is critical. Children 5
porous surfaces should be scrubbed with disinfectant using years of age or younger should be kept out of patient care areas.
brushes, and then rinsed with water after the contact time has Strict attention to hygiene and protective apparel is necessary
elapsed. when handling soiled cat litter or animal feces, because of the
When hospital surfaces are cleaned, attention should be paid risk for transmission of enteropathogens. The reader is referred
to corners, under cabinets, chairs, the bases of examination to other chapters in this book for public health implications of
tables, door handles, elevator buttons, shelves, sinks, faucets, specific infectious diseases that may be encountered in small ani-
and other surfaces that might otherwise be ignored. Personal mal hospitals. If possible, small animal clinics should establish
items such as cell phones and stethoscope heads should be regu- a relationship with a doctor who is prepared to consult on zoo-
larly disinfected with disinfectant wipes. Cell phones should be notic exposures with staff members.
disinfected at least daily, and stethoscope heads wiped between
patients. Stethoscope tubing should be cleaned regularly with
soap and water. Disinfection of other fomites should be per- Transmissible Disease Surveillance
formed on a regular (at least daily) basis. These include digital and Reporting
thermometers, computer keyboards and mice, land-line tele-
phones, calculators, microscopes, otoscopes, and blood pres- A transmissible disease surveillance program allows collection
sure cuffs. of baseline data that establishes the prevalence of certain infec-
Cleaning staff should wear gloves when general hospital tious diseases, so that possible outbreak situations can be readily
disinfection is performed. This may not be necessary for spot identified. It can also provide background prevalence informa-
cleaning of areas such as examination room tables or fomites. tion on antimicrobial drug susceptibility patterns, which can
Depending on the disinfectant, additional protective attire, such assist in the initial selection of antibacterial drugs for individual
as a gown, boots, or face mask, may be required if there is a animal patients while culture and susceptibility test results are
probability of significant splashing during the disinfection pro- in progress. Information is generally collected for multidrug-
cess. After disinfection is complete, protective clothing should resistant bacterial infections, zoonotic diseases, highly conta-
be removed and handwashing performed. The individuals gious diseases, pathogens that are difficult to inactivate with
CHAPTER 11  Infection Control Programs for Dogs and Cats 115

disinfectants, or agents of regulatory concern. Although surveil-


lance may seem like a difficult, time-consuming, and expensive BOX 11-3
measure, in reality, it can be easy and cost-effective and can
provide important information.
Infectious Diseases of Dogs and Cats That Are Reportable or
There are two main forms of infection control surveillance Potentially Reportable to Public Health Agencies
applicable to veterinary hospitals: active and passive. Active
surveillance involves collection of data specifically for infection Amebiasis Q fever
control purposes. This can provide the highest quality and most Granulocytic anaplasmosis Rabies
relevant information, but it can be expensive and time consum- Brucellosis Salmonellosis
ing. Examples of this would be collection of swab specimens Campylobacteriosis Tularemia
to screen for infection with methicillin-resistant Staphylococ- Coccidioidomycosis Tuberculosis
cus pseudintermedius (MRSP) from dogs before surgery as part Cryptococcus gattii MRSA (not MRSP)
of MRSP outbreak investigation. Active surveillance is a core infection Novel H1N1 influenza
component of infection control in most human hospitals, but Cryptosporidiosis virus infections
it is only sporadically used in veterinary medicine and is rarely Giardiasis West Nile Virus infection
needed in most veterinary clinics. It is typically reserved for Leishmaniosis Yersinia infections
large facilities with increased infection control risks and per- Leptospirosis
sonnel available to direct such testing, or as a part of outbreak Lyme disease
control.
In contrast, passive surveillance is a practical, easy, and
cost-effective surveillance approach that can and should be per-
formed in every veterinary clinic. It involves the use of data that Environmental cultures are rarely informative and so are not
are already available, such as information about surgical site considered a useful routine infection control tool. They may
infections collected during routine follow-up or culture results be used to detect a specific pathogen if an outbreak of an HAI
from clinical testing. The quality of passive surveillance data is suspected when there is suspicion that the environment is a
can be limited by poor or incomplete record keeping or sporadic source of exposure. However, it is often difficult to distinguish
use of appropriate diagnostic tests, but if properly collected and cause from effect (i.e., environmental contamination that leads
if potential biases are understood, passive surveillance data can to transmission vs. environmental contamination that occurs as
provide important insight into aspects such as endemic disease a result of contamination from a patient in the absence of a risk
rates, common pathogens, and antimicrobial susceptibility of transmission).
trends. To facilitate passive surveillance, clinicians should be Although surveillance programs require time, effort, and
encouraged to use appropriate diagnostic testing to determine expense, in the long term they may save morbidity and mortal-
the etiology of nosocomial infections, even if the clinical con- ity and reduce costs that relate to control of large outbreak situ-
sequences are not severe. They also should be encouraged to ations or the legal ramifications of HAIs.
confirm a diagnosis in animals with suspected transmissible dis- When zoonotic diseases are identified, all in-contact individ-
ease. This allows clients to protect their other animals and their uals and, if necessary, public health authorities should be noti-
families and friends who might be in contact with the pet, and fied in the appropriate manner. The specific diseases that must
it benefits the hospital. be reported to authorities vary among geographic locations.
Another form of surveillance that is easy to perform and Examples of diseases that may be of interest to public health
potentially very useful is syndromic surveillance. This involves authorities are listed in Box 11-3.
surveillance for readily identifiable syndromes (i.e., diarrhea,
cough) instead of specific diagnoses. Although syndromes do not Surgical Preparation
indicate a specific disease, they can indicate an increased risk of
infectious disease. Syndromic surveillance is an initial screening Surgical site infections (SSIs) are an uncommon but important
tool that can be used by all personnel, including lay personnel, and sometimes devastating complication of surgery. Every
to flag potentially high-risk cases and allow for early implemen- patient undergoing a surgical procedure is at some risk of SSI.
tation of enhanced infection control practices. All clinic person- Standard practices have been developed to reduce the risk of
nel should be made aware of certain syndromes that indicate SSIs. Although a wide range of practices are relevant, prepara-
the need either for isolation or for further investigation, such tion of the patient and preparation of the surgeon are critical.
as diarrhea, fever of unknown origin, acute neurologic disease,
wound infections, and acute respiratory tract disease. Protocols Patient Preparation
to deal with these animals on arrival should be developed. As The patient’s endogenous microflora is an important source of
an example, a dog with an acute onset of cough should be con- pathogens that cause SSIs. Careful preparation of the patient,
sidered potentially infectious, and if front office personnel note therefore, can help reduce contamination of the surgical site
this syndrome at the time the appointment is made, the dog during surgery. The goal of preoperative surgical site manage-
can be handled appropriately on arrival (e.g., admitted directly ment is to eliminate potential pathogens while not creating a
to isolation or an examination room, with personnel wearing physical environment that is more conducive to bacterial colo-
enhanced barrier precautions from the onset). The role of lay nization or infection postoperatively. Bathing of the patient pre-
staff (i.e., front office stall) is critical, as these people are the operatively is reasonable if there is significant contamination of
ones who are most able to identify such cases before they enter the haircoat19 and if the patient’s coat can be dried by the time
the clinic and ensure that they are properly handled to prevent of surgery. In most situations, bathing is not required. Rather,
nosocomial or zoonotic transmission. careful hair removal and skin antisepsis are the main measures.
116 SECTION 3  Basic Principles for Infection Control

The goals of surgical scrubbing of patients are to reduce bacte-


rial counts, reduce debris, and facilitate later antisepsis. Scrub- BOX 11-4
bing should be done as atraumatically as possible. Minimizing
skin damage during clipping and scrubbing is essential, because
Example of Criteria Specified within a Hospital
skin damage from excessive attempts to clip all remaining for Use of Restricted Antimicrobial Drugs
pieces of hair or from forceful scrubbing of the surgical site
can create an environment that is more amenable to bacterial • Infection must be documented based on clinical abnor-
growth. This predisposes to infection rather than reducing the malities and culture.
risk. Clipping should be performed after anesthesia, to reduce • Subclinical infections should not be treated with these
the risk of trauma associated with a struggling patient and antibiotics. These antibiotics should be reserved for
to minimize the time between clipping and surgery. Clipping infections that are life threatening.
should be done outside the operating environment. There is cur- • Resistance to all other reasonable options and suscepti-
rently no information that relates to optimal methods of clean- bility to the chosen antimicrobial must be documented.
ing and disinfecting clippers. Repeated use of clipper blades • The infection must be potentially treatable.
without sterilization not surprisingly results in higher levels of • The clinician should contact the infectious disease
bacterial contamination of blades20; however, the clinical rel- control officer or a clinical microbiology faculty
evance of this is unclear, because the surgical site is cleaned member by email or telephone to discuss antimicrobial
and disinfected after clipping. Regular cleaning and disinfection susceptibility test results, determine whether there are
of clippers are probably useful, and they should be thoroughly any other viable options, and confirm that a restricted
cleaned and disinfected after use on an animal with a poten- antimicrobial is necessary.
tially transmissible infection (e.g., an animal with diarrhea), on
an area where the skin is broken, or on any area where the skin
or hair is significantly contaminated with feces, urine, blood, or
other body fluids. Regardless of the method used, a thorough handwash with
After hair removal, various approaches for skin antisepsis careful cleaning under the fingernails must be performed at the
can be used. There is little outcome-based evidence of the rela- beginning of each day.23 Long (>1⁄4-inch) and artificial nails are
tive efficacy of different approaches in the human literature. prohibited in many human health care facilities and some vet-
Typically, a three-step process is used, with initial scrubbing erinary hospitals because they harbor pathogenic bacteria24 and
of the site with biocidal (i.e., chlorhexidine, povidone-iodine) are associated with surgical glove tears.
soap, followed by application of alcohol for biocidal effects and A cap and mask must be worn during hand antisepsis, and
to remove oils, and a final application of a biocidal solution sterile gown and gloves must then be donned using appropriate
with residual activity (e.g., chlorhexidine). technique.

Surgeon Preparation Antimicrobial Drug Use


The surgeon’s body and clothing are potential sources of SSI
pathogens, and standard practices have been developed to The use of antimicrobial drugs contributes to the selection for
reduce the risk of contamination of the surgical site. One aspect antimicrobial-resistant bacteria. Inclusion of a policy on anti-
is the use of proper protective clothing. Every person in the microbial drug use in the infection control protocol may help
operating environment should wear clean surgical scrubs. These to reduce the prevalence of multidrug-resistant HAIs. On their
scrubs should be dedicated for use only in the operating room own, fever and leukocytosis are not justification for antimicro-
or should be covered with a clean laboratory coat whenever the bial drug treatment. Whenever possible, when infection is sus-
individual is outside of the operating room. pected, attempts to obtain material for culture and susceptibility
Surgical hand antisepsis is critical because of the close con- testing should be made before antimicrobial drug treatment is
tact of the hands with the surgical site and the relatively high initiated. When secondary bacterial infection is likely, attempts
incidence of grossly evident breaks or micro-breaks in gloves. to identify and treat the underlying cause should be made. In
Surgical hand antisepsis is designed to greatly reduce bacte- an effort to minimize the emergence of resistance in bacterial
rial burdens on the hands, particularly the abundant transient flora of animals in the hospital and decrease the risk of resis-
microflora that contains most of the relevant pathogens that tant HAIs, the use of several antimicrobials (e.g., certain par-
cause SSIs. Surgical antisepsis must find a balance between enteral third-generation cephalosporins, vancomycin, linezolid,
effective elimination of pathogens, minimization of trauma to and carbapenems such as imipenem and meropenem) may be
the skin (because skin irritation facilitates bacterial growth), restricted unless criteria must be met (Box 11-4).
and time constraints. The traditional approach to surgical hand
antisepsis has involved structured scrubbing of the hands for a
predetermined time. Recommended scrub times vary between Prevention of Infectious Disease in Shelters
products but are typically 2 to 4 minutes. and Breeding and Boarding Facilities
Application of alcohol-chlorhexidine combinations has been
evaluated as a replacement for surgical scrubbing and has been Attention to hand hygiene and proper disinfection of fomites
shown to be more effective than standard surgical scrub meth- and the environment as outlined for hospitals can also be used
ods.21,22 Specific manufacturer instructions should be followed, to reduce transmission of infectious pathogens in shelters and
and hands and arms must be dry before application of gloves. in breeding and boarding facilities (Box 11-5). Disinfectants
The use of alcohol-based surgical hand antisepsis products is with activity against parvoviruses and feline calicivirus should
encouraged as a replacement for traditional scrubbing. be used routinely, such as a 1:30 dilution of household bleach,
CHAPTER 11  Infection Control Programs for Dogs and Cats 117

separate airflow from areas that house healthy animals. Mass


BOX 11-5 treatment of dogs and cats with upper respiratory tract disease
with antimicrobials without attention to the underlying causes
Factors That Should Be Considered for the Reduction of only results in selection for antimicrobial drug resistant organ-
Infectious Disease Transmission in Animal Housing Facilities isms and is not recommended. Detailed information on the con-
trol of infectious diseases in shelter environments is beyond the
Adequate ventilation and air quality scope of this book but can be found elsewhere.25 The American
Adequate temperature Association of Shelter Veterinarians and the Humane Society
Adequate lighting conditions (including provision of dark- of the United States have published guidelines for operation of
ness at night) animal shelters.26,27
Population density/space per animal
Daily removal of fecal and urine contamination and SUGGESTED READINGS
disinfection Association of Shelter Veterinarians. Guidelines for standards of care
Adequate drainage in animal shelters. 2010. oacu.od.nih.gov/disaster/ShelterGuide.pdf.
Separate housing areas for dogs and cats Last accessed November 17, 2012.
Isolation for sick animals Boyce JM, Pittet D. Guideline for hand hygiene in health-care set-
Separate housing areas for young and adult animals tings. MMWR. 2002;51. http://www.cdc.gov/handhygiene/Guideli
Individual housing or small groups of two to four compat- nes.html. Last Accessed November 17, 2012.
Canadian Committee on Antibiotic Resistance. Infection
ible animals
prevention and control best practices for small animal veterinary
Separation of elimination, feeding, and resting areas
clinics. 2008. http://www.wormsandgermsblog.com/2008/04/promo/
Elevated resting areas services/infection-prevention-and-control-best-practices-for-small-
Elevated cages for cats animal-veterinary-clinics/. Last accessed November 17, 2012.
Provision of hiding areas WHO guidelines on hand hygiene in health care. http://www.cdc.gov/
Sound control handhygiene/guidelines.html. Last accessed November 17, 2012.
Use of surfaces that can be readily disinfected
Proper use of appropriate disinfectants REFERENCES
Hand hygiene and protective clothing
1. Edmond EB, Wenzel RP. Isolation. In: Mandell GL, Bennett JE,
Fomite control
Dolin R, eds. Principles and Practice of Infectious Diseases. 7th ed.
Use of appropriate vaccination protocols
Philadephia, PA: Elsevier; 2010:3673-3676.
Order of care (young animals, healthy adults, and then 2. Longtin Y, Sax H, Allegranzi B, et al. Hand hygiene. N Engl J Med.
sick animals) 2011:e24.
Proper nutrition 3. WHO guidelines on hand hygiene in health care. 2009. http://
Pain management www.cdc.gov/handhygiene/Guidelines.html. Last accessed Novem-
Free access to clean water ber 17, 2012.
Rodent and pest control 4. Guideline for hand hygiene in health-care settings. 2002. http://
Parasite control www.cdc.gov/handhygiene/Guidelines.html. Last accessed Novem-
Daily monitoring by properly trained individuals ber 17, 2012.
5. McGovern B, Doyle E, Fenelon LE, et al. The necktie as a potential
Proper diagnosis of disease outbreaks
vector of infection: are doctors happy to do without? J Hosp Infect.
2010;75:138-139.
6. Centers for Disease Control and Prevention. Guideline for the pre-
vention of surgical site infection. 1999. http://www.cdc.gov/HAI/
potassium peroxymonosulfate, or accelerated hydrogen perox- ssi/ssi.html. Last accessed November 17, 2012.
ide solutions. Removal of organic matter, the use of surfaces 7. Lenz J, Joffe D, Kauffman M, et al. Perceptions, practices, and con-
that can be adequately disinfected, and proper contact times are sequences associated with foodborne pathogens and the feeding of
essential. raw meat to dogs. Can Vet J. 2009;50:637-643.
8. Guideline for isolation precautions: preventing transmission of
Factors that increase stress should also be minimized, such
infectious agents in healthcare settings. 2008. http://www.cdc.gov/
as high population densities, the grouping of animals, and poor
hicpac/2007ip/2007isolationprecautions.html. Last accessed Novem-
nutrition. Cats and dogs should be separated from one another. ber 17, 2012.
In cats, the use of methods that reduce stress, such as provision 9. Rutala WA, Weber DJ. Disinfection, sterilization, and control of
of a low-stress cage environment, can dramatically reduce the hospital waste. In: Mandell GL, Bennett JE, Dolin R, eds. Principles
frequency of upper respiratory tract disease. Compartmental- and Practice of Infectious Diseases. 7th ed. Philadephia, PA: Elsevier;
ized housing allows animals to urinate and defecate away from 2010:3677-3695.
areas where they rest and eat, and provision of a hiding area so 10. Akamatsu T, Minemoto M, Uyeda M. Evaluation of the antimicro-
that animals can retreat from visual stimulation can also reduce bial activity and materials compatibility of orthophthalaldehyde as
stress. A total of 10 to 20 air changes per hour is often rec- a high-level disinfectant. J Int Med Res. 2005;33:178-187.
11. Hession SM. Endoscope disinfection by ortho-phthalaldehyde
ommended for animal housing, but higher levels of ventilation
in a clinical setting: an evaluation of reprocessing time and costs
may be needed with increased population density and concen-
compared with glutaraldehyde. Gastroenterol Nurs. 2003;26:
tration of airborne contaminants. Cages should be cleaned and 110-114.
disinfected at least daily. The use of compartmentalized housing 12. Cooke RP, Goddard SV, Whymant-Morris A, et al. An evaluation
areas also facilitates cleaning and disinfection without requir- of Cidex OPA (0.55% ortho-phthalaldehyde) as an alternative
ing an animal to be removed from the housing area. Isolation to 2% glutaraldehyde for high-level disinfection of endoscopes.
areas for sick animals should be present and these should have J Hosp Infect. 2003;54:226-231.
118 SECTION 3  Basic Principles for Infection Control

13. Omidbakhsh N. Evaluation of sporicidal activities of selected 20. Masterson TM, Rodeheaver GT, Morgan RF, et al. Bacteriologic
environmental surface disinfectants: carrier tests with the spores evaluation of electric clippers for surgical hair removal. Am J Surg.
of Clostridium difficile and its surrogates. Am J Infect Control. 1984;148:301-302.
2010;38:718-722. 21. Mulberrry G, Snyder AT, Heilman J, et al. Evaluation of a water-
14. Alfa MJ, Lo E, Wald A, et al. Improved eradication of Clostridium less, scrubless chlorhexidine gluconate/ethanol surgical scrub for
difficile spores from toilets of hospitalized patients using an acceler- antimicrobial efficacy. Am J Infect Control. 2001;29:377-382.
ated hydrogen peroxide as the cleaning agent. BMC Infect Dis. 2010; 22. Hobson DW, Woller W, Anderson L, et  al. Development and
10:268. evaluation of a new alcohol-based surgical hand scrub formulation
15. Howie R, Alfa MJ, Coombs K. Survival of enveloped and non- with persistent antimicrobial characteristics and brushless applica-
enveloped viruses on surfaces compared with other micro-organ- tion. Am J Infect Control. 1998;26:507-512.
isms and impact of suboptimal disinfectant exposure. J Hosp 23. Larson EL. APIC guideline for handwashing and hand antisepsis in
Infect. 2008;69:368-376. health care settings. Am J Infect Control. 1995;23:251-269.
16. Eleraky NZ, Potgieter LN, Kennedy MA. Virucidal efficacy of four 24. Pottinger J, Burns S, Manske C. Bacterial carriage by artificial ver-
new disinfectants. J Am Anim Hosp Assoc. 2002;38:231-234. sus natural nails. Am J Infect Control. 1989;17:340-344.
17. Weber DJ, Rutala WA, Sickbert-Bennett EE. Outbreaks associated 25. Miller L, Hurley KF. Infectious Disease Management in Animal
with contaminated antiseptics and disinfectants. Antimicrob Agents Shelters. Ames, IA: Wiley-Blackwell; 2009.
Chemother. 2007;51:4217-4224. 26. Humane Society of the United States. HSUS guidelines for stan-
18. Maki DG, Ringer M, Alvarado CJ. Prospective randomised trial dard operating procedures for animal shelters. http://dev.animal
of povidone-iodine, alcohol, and chlorhexidine for prevention of sheltering.pub30.convio.net/resource_library/policies_and_
infection associated with central venous and arterial catheters. Lan- guidelines/guidelines_for_standard_operating_procedures.html.
cet. 1991;338:339-343. Last accessed November 17, 2012.
19. Stick JA. Preparation of the surgical patient, the surgery facility, 27. Association of Shelter Veterinarians. Guidelines for standards
and the operating team. In: Auer JA, Stick JA, eds. Equine Surgery. of care in animal shelters. 2010. oacu.od.nih.gov/disaster/Shelter
3rd ed. Philadelphia, PA: Saunders Elsevier; 2006:123-140. Guide.pdf. Last accessed November 17, 2012.
CHAPTER 12

Immunization
Jane E. Sykes

w KEY POINTS
• A  ctive immunization can partially or completely protect dogs • O  ther adverse effects of vaccine administration are uncommon
and cats from severe consequences of infection with a variety to rare but include hypersensitivity reactions, disease induced
of different pathogens, and in some cases it reduces shedding by live attenuated vaccine organisms, and injection-site sarco-
of these pathogens. mas in cats.
• Vaccines contain attenuated live microorganisms, inactivated • The decision to administer a vaccine should be based on dis-
microorganisms, or portions of these organisms. They also con- cussion of risks and benefits between the veterinarian and pet
tain preservatives and adjuvants. owner. This should be documented in the medical record.
• Failure of immunization can occur with improper storage or • Guidelines for vaccine selection and administration have been
administration of vaccines, a large challenge dose, host factors published by a number of veterinary bodies, such as the AAFP,
such as concurrent infections or disease, and interference by AAHA, AVMA, and WSAVA; suggestions can also be found in
maternal antibody. Appendix I.

INTRODUCTION Vaccine Composition and Types of Vaccines


Immunization refers to artificial induction of immunity or protec- A vaccine is a suspension of attenuated live or inactivated
tion from infectious disease and may be active or passive. Active microorganisms, or parts thereof, that is administered to induce
immunization involves administration of vaccines that stimulate immunity. In addition to protective antigens, vaccines may
cell-mediated or humoral immunity, or both, to a specific patho- contain preservatives and stabilizers as well as specific antibi-
gen. Passive immunization refers to the administration of antibod- otics to preserve the antigen and inhibit bacterial and fungal
ies in order to provide temporary protection from disease and can growth within the vaccine. Some vaccines also contain an adju-
occur through acquisition of maternally derived antibody (MDA) vant to enhance the immune response to the antigen. Although
transplacentally, in colostrum, or milk; or treatment with prepara- the mechanisms are not completely clear, adjuvants can delay
tions that contain specific or nonspecific immunoglobulins (see the release of antigen from the site of injection and induce the
Immunomodulators, Chapter 7, and post-exposure prophylaxis for secretion of chemokines by leukocytes.4 The most widely used
rabies, Chapter 13). Readers are referred to advanced immunology adjuvants are particulate adjuvants, such as those that contain
texts for detailed descriptions of the physiology of active and pas- aluminum salt precipitates such as aluminum hydroxide.5 Other
sive immunity.1 particulate adjuvants include immunostimulators such as sapo-
The goal of immunization is to generate a protective immune nin, which is present in a canine Leishmania vaccine.
response of prolonged duration against a specific infectious dis- Attenuated live vaccines (or modified live vaccines) con-
ease, with minimal adverse effects. Because of the potential for tain microorganisms that are artificially manipulated so as to
adverse effects, vaccination should be performed only if there negate or greatly reduce their virulence, or are field strains of
is a risk for significant morbidity or mortality from an infec- low virulence. Repeated passage through cell culture is the most
tious disease. Since the 1950s, a huge number of vaccines for common means of attenuation. Because they replicate in the
dogs and cats have been developed and marketed worldwide, host, organisms in attenuated live vaccines usually stimulate an
and more are in development. Nevertheless, it is estimated that immune response that most closely mimics the protection that
even in developed countries such as the United States, only 30% results from natural infection. Vaccination with attenuated live
to 50% of dogs are properly immunized, and possibly an even canine parvovirus (CPV) and canine distemper virus (CDV) vac-
smaller proportion of cats.2,3 Appropriate vaccination of a larger cines in the absence of MDA can result in protective immune
proportion of the pet population may assist in reduction of the responses within 3 days of a single injection, which may be
prevalence of infectious diseases through the induction of herd followed by immunity that lasts many years, if not for life.6-8
immunity. Partial immunity after vaccination with attenuated live CDV
With the appearance of injection-site sarcomas in cats, increased and feline panleukopenia virus (FPV) vaccines can occur within
emphasis has been placed on vaccine safety, and a change from hours.3,9,10 In addition, vaccine organisms that are shed can
annual to 3-yearly immunization protocols for some vaccines has serve to immunize other animals in a population. However, the
been recommended, with administration of other vaccines based potential for reversion to virulence or vaccine-induced disease
on exposure risk. Vaccines have had a profound influence in the exists. Vaccine-induced disease is most likely to occur in highly
control of infectious disease, and for many vaccines the benefits of immunosuppressed animals. Attenuated live vaccines also
vaccination outweigh the risks. have the potential to cause some immunosuppression in their

119
120 SECTION 3  Basic Principles for Infection Control

TABLE 12-1
Advantages and Disadvantages of Attenuated Live and Inactivated Vaccines
Attenuated Live Inactivated
Advantages Rapid onset of immunity Safe, even in immunocompromised and pregnant
Sustained immunity after single dose animals
May immunize others in populations Do not interfere with development of ­immunity
Improved breakthrough of maternal antibody from other vaccines
interference Stable in storage
Disadvantages Potential for reversion to virulence Slow onset of immunity
Virulence in the immunocompromised Multiple boosters required
Contraindicated in pregnancy Often highly adjuvanted, with greater potential
May cause immune suppression for adverse effects
Can interfere with development of immunity if administered Reduced degree of protection compared with at-
within days to 2 weeks of another vaccine tenuated live vaccines
Less stable in storage Poor breakthrough of maternal antibody
Potential for vaccine contamination ­interference

own right,11,12 or they may shift the balance from Th1 to Th2 Subunit vaccines contain specific structural components of a
immune responses.13 Rarely, this can lead to clinical disease. microbe that stimulate a protective immune response, together
For example, an outbreak of salmonellosis was reported in cats with adjuvant. They contain reduced amounts of foreign protein,
after use of a high-titered attenuated live FPV vaccine.14 Very which minimizes the potential for hypersensitivity reactions.
rarely, contamination of attenuated live vaccines has occurred Recombinant DNA vaccines are created through manipu-
with other pathogenic microorganisms present within cell cul- lation of the DNA of a pathogen in the laboratory, with the
tures used to propagate the vaccine. negation of pathogen virulence. Sometimes this also can allow
Generally speaking, inactivated vaccines are less effective diagnostic tests to differentiate naturally infected from vacci-
than attenuated vaccines, because replication in the host does nated animals (DIVA), because of differences in the antibody
not occur. They produce weaker immune responses of shorter response evoked by the vaccine. There are several different types
duration, and more frequent booster immunizations may be of recombinant DNA vaccines:
required. Two initial doses of vaccine 3 to 4 weeks apart are 1. Recombinant subunit vaccines. These are produced by
essential to produce an effective immune response, and if more cloning one or more genes for a protective antigen into an
than 6 weeks elapses between these doses, it has been recom- expression vector, such as in Escherichia coli. The protein
mended that the series should be repeated.15 Beyond the initial expressed by the bacteria is then purified and used in the vac-
vaccination series, it is not clear whether lapsed annual boosters cine ­(Figure 12-1, A). An example of a recombinant subunit
require the series to be restarted. This is not considered neces- vaccine is the Lyme recombinant OspA vaccine.
sary for human immunization16 but has been suggested for dogs 2. Deletion mutant vaccines. These are produced by deleting
when more than 2 or 3 years elapses between boosters.15 Inacti- virulence genes from a pathogen while protective antigens
vated vaccines usually contain adjuvant as well as a large infec- are left in place. There are currently no such vaccines for
tious dose to improve immunogenicity. They are safer than live dogs and cats.
attenuated vaccines for use during pregnancy and in very young 3. Vectored vaccines. These are produced by inserting genes for
or debilitated animals. Although bacterins have traditionally one or more protective antigens into the genome of a virus.
been associated with a greater likelihood of allergic reactions The virus replicates in the host and expresses the antigens
than live attenuated vaccines, newer inactivated vaccines are but is nonpathogenic (see Figure 12-1, B). Currently avail-
safer and have reaction rates that more closely approach those able vectored vaccines for dogs and cats use canarypox virus
of live attenuated vaccines. The maximum duration of immunity as a vector.
that is induced by commercially available bacterins for dogs and 4. DNA vaccines. These consist of naked DNA that encodes
cats remains largely unknown, partly because challenge studies the antigens required for protective immunity. The DNA is
that evaluate long-term duration of immunity are prohibitively injected directly to the animal using an inoculation system.
expensive. However, some inactivated viral vaccines have been The DNA is then taken up by host cells and translated into
shown to have durations of immunity in excess of 7 years in antigen. Both humoral and cell-mediated immune responses
cats.17 Caution is required when extrapolation is made from are produced. DNA vaccines are not currently available
the duration of immunity for one product to that for a similar commercially for use in dogs and cats.
product from a different manufacturer, because it may not be
equivalent. Although bacterins usually do not protect all animals
from infection, they may prevent clinical illness. In some cases, Vaccine Storage, Handling,
natural infection of vaccinated animals serves to further boost and Administration
the immune response, and this can influence duration of immu-
nity in the field. The advantages and disadvantages of attenuated Vaccines should be stored and administered according to label
live and inactivated vaccines are shown in Table 12-1. recommendations. Inactivation of vaccines can occur if they
CHAPTER 12  Immunization 121

A B

FIGURE 12-1  Examples of recombinant DNA vaccines. A, Recombinant subunit vaccine. The gene of interest is inserted into an expression vector such as a plasmid taken up by
Escherichia coli, which subsequently produces large amounts of an immunogenic protein. This is purified and used in the vaccine. B, Vectored vaccine. The gene or genes of interest are
inserted into a canarypox or vaccinia vector, which is then inoculated into an animal. Replication of the vector within the host is followed by expression of the immunogenic protein.

are inadvertently frozen or heated to excessive temperatures, 4 weeks is preferred for human patients.16,21 Inactivated vac-
exposed to excessive amounts of light, or used beyond their cines do not produce interference in this way.16 If possible,
expiration date. Hands should be washed before preparation administration of vaccines to animals that are under anesthesia
and administration of the vaccine. Lyophilized products should should be avoided because adverse reactions may be difficult
be reconstituted with the proper diluent, and different vaccines or impossible to recognize in this situation. It is not necessary
should not be mixed in the same syringe or vial. Reconstituted to re-administer an intranasal vaccine if the animal coughs or
products should be used immediately. It has been recommended sneezes after administration.
that attenuated live vaccines be discarded if more than 1 hour The site and route of administration, product, serial number,
has lapsed since reconstitution,15 although no published reports expiry date, and individual who administered the vaccine should
exist of the viability of vaccine organisms over time after recon- be recorded for each vaccine administered.2 Vaccine vials often
stitution or of the ability of stored, reconstituted vaccine to elicit possess adhesive labels that can be easily removed and applied
an immune response. Vaccines should only be used in the ani- to a paper medical record.
mal species for which they are labeled, or serious adverse effects
or failure of immunization can occur. Components of the Immune Response
If vaccines for multiple different pathogens are to be admin-
istered simultaneously, they should be injected at distant sites The immune response is divided into innate and adaptive
or, if possible, a combination vaccine should be used. Simul- immune responses. The innate immune response is nonspecific
taneous vaccination for more than one pathogen does not and acts as an immediate line of defense against an infection.
appear to interfere with immune responses to each component Components of the innate immune response consist of natu-
of the vaccine,18-20 and vaccine manufacturers must demon- ral killer cells, which recognize host cells that are infected by
strate that the protection that occurs for a specific pathogen viruses; complement, which is activated by bacterial cell wall
after vaccination with a combination product equals the pro- components; and phagocytes, such as macrophages and den-
tection that occurs when a vaccine for only that pathogen is dritic cells. The adaptive immune response develops over sev-
given. In contrast, successive parenteral administration of dif- eral days and involves presentation of antigen by dendritic
ferent attenuated live vaccines at 3 to 14 day intervals has the cells in association with the major histocompatibility complex
potential to interfere with immune responses. An interval of and stimulation of B and T cell responses, together with the
122 SECTION 3  Basic Principles for Infection Control

rabies vaccines than large-breed dogs.25 Administration of vac-


BOX 12-1 cines to febrile animals or animals with moderate to severe ill-
ness should be avoided if possible until recovery has occurred,
Factors That Can Affect Immune Responses to Vaccines because the immune response to the vaccine may be suboptimal.
Failure of immunization can result from an inadequate dose
Target pathogen (e.g., respiratory versus systemic pathogen) of antigen. Thus, division of a single vaccine dose for adminis-
Vaccine composition (e.g., inadequate adjuvant) tration to a larger number of dogs and cats, or small-breed dogs
Route of administration as opposed to large-breed dogs, may lead to failure of immuni-
Young age zation. Veterinarians should not split vaccine doses because this
Breed/genetic factors shifts the liability from the vaccine manufacturer to the veteri-
Nutrition narian if vaccine failure occurs. Immunization can also fail in
Pregnancy status the face of an overwhelming challenge dose.
Concurrent moderate to severe illness The route of administration can influence the type of immune
Fever response generated. Subcutaneous administration is associated
Immunosuppressive drugs primarily with an IgG response, and rarely induces high levels of
Presence of maternal antibody secretory IgA antibodies. In contrast, intranasal administration
Improper vaccine storage and administration results in an IgA and, to a lesser extent, an IgG response. Immu-
Vaccination with an attenuated live viral vaccine within nogenicity and safety may be compromised when a vaccine is
last 3 days to 2 weeks administered using the incorrect route.
Inadequate time allowed for immunization before expo- In young animals, MDA can neutralize vaccine antigens and
sure to field organisms interfere with effective immunization. This is one of the most
common reasons for vaccine failure in dogs and cats. Any MDA
titer against CPV has the potential to interfere with immuniza-
formation of memory B cells. The nature of the innate response tion. The amount of MDA in a puppy or kitten at any one point
influences the subsequent adaptive response. Cells of the innate in time cannot be predicted because it varies depending on the
immune system possess pattern recognition receptors that can titer of the dam and the amount of colostrum ingested after
recognize patterns that are characteristic for various pathogens birth. As a result, a series of vaccinations are administered in
(pathogen-associated molecular patterns, or PAMPs), including order to increase the chance that successful immunization will
Toll-like receptors and NOD-like receptors. PAMPs are under occur soon after the decline of MDA titers to sufficiently low
investigation for use as adjuvants in human and animal vaccines concentrations (Figure 12-2). Nevertheless, a window always
in order to create improved T cell immune responses.4,22 exists when MDA concentrations are high enough to interfere
with immunization, but not sufficient to prevent natural infec-
Determinants of Immunogenicity tion. This window is known as the window of susceptibility or
the window of vulnerability. The use of recombinant vectored
All vaccines that are available for dogs and cats induce cell- vaccines can overcome the interference by MDA, although the
mediated immunity (CMI) with induction of immunologic mem- extent to which this applies in animals that have passive immu-
ory and a booster effect on repeat administration. Although the nity to the vector virus (i.e., immunity transferred from a dam
presence of antibody correlates with protection for some patho- that was immunized with a recombinant vector vaccine) requires
gens, such as CDV and CPV, a lack of antibody does not infer clarification. Because replication of the vector is aborted, the
a lack of protection, because of the presence of CMI, which is immune response to the vector itself may be reduced. As a result,
more difficult to measure. passive transfer of neutralizing antibody titers to the vector may
Vaccines rarely protect all vaccinated individuals from infec- not occur. Mucosal vaccines can also provide greater protec-
tion and disease. In particular, vaccines for canine and feline tion in the face of MDA; the mucosal immune system matures
respiratory pathogens do not prevent disease but can reduce the shortly after birth.26,27
prevalence and severity of disease as well as reduce the number Whenever possible, animals should be isolated until sufficient
of organisms shed. Limited immunity following vaccination is time has elapsed for proper immunization. For most parenteral
especially likely for infections for which immunity after natural and mucosal vaccines, this is 1 week (and at the absolute mini-
infection is partial or short-lived. mum, 3 days) after inoculation. Vaccine failure can also occur
The ability of a vaccine to induce an immune response in animals that are incubating the disease for which vaccination
depends not only on the target pathogen, vaccine composition, is performed at the time of vaccination.
and route of administration, but also on host factors such as
age, nutrition, pregnancy status, stress, concurrent infections, Measurement of the Immune Response
and immune status, including the presence or absence of pas-
sively acquired antibody (Box 12-1). Some of these factors may For some vaccines, such as rabies, CDV, CPV, and FPV, the
also influence vaccine safety. Some animals, particularly dogs of presence of circulating antibodies correlates with protection
the Rottweiler breed, may have an impaired ability to respond (Table 12-2). Thus, serologic assays have been used in dogs and
to vaccination. These dogs have been termed nonresponders.2,23 cats to decide whether vaccination is necessary or likely to be
This situation is probably rare if efficacious vaccines are used effective. These serologic assays have also been used to clear
and booster vaccines are administered. Young dogs, less than pets for travel.
1 year of age, have a significantly reduced response to vacci- Although tests that measure antibody responses in dogs
nation with rabies virus vaccines when compared with adult and cats have improved in recent years, different laboratories
dogs.24 Small-breed dogs have a greater serologic response to can report significantly different values for the same serum
CHAPTER 12  Immunization 123

FIGURE 12-2  Influence of maternal antibody (MDA) on immunization. Puppies and kittens acquire variable amounts of MDA transplacentally and through colostrum after birth. This
binds to vaccine antigens and inhibits the immune response. A series of vaccines are administered to maximize the chance of inducing an immune response as MDA concentrations decline.
The window of susceptibility is the period of time when MDA concentrations are high enough to interfere with immunization, but not sufficient to prevent natural infection. High antigen
mass vaccines provide protection earlier than low mass vaccines. (From Greene CE, Schultz RD. Immunoprophylaxis. In: Greene CE, ed. Infectious Diseases of the Dog and Cat, 3 ed. St Louis,
MO: Saunders; 2006.)

to develop disease after challenge, possibly because of over-


TABLE 12-2 whelming exposure or immune suppression. Measurement of
Antibody Titers That Correlate with Protection against antibody titers may be considered for animals that have had
Distemper, Parvovirus, and Rabies previous adverse responses to vaccination, particularly suscep-
tible breeds (e.g., Rottweilers and CPV infection). The World
Minimum Small Animal Veterinary Association (WSAVA) has suggested
Pathogen Protective Titer Methodology Used that puppies could be tested at least 2 weeks after the final
puppy vaccine to decide whether further vaccination for CDV
Canine distemper ≥1:16 to 1:20 Serum neutralization or CPV is necessary.2 Negative titers should prompt additional
virus (SN) vaccination for these puppies.
Canine parvovirus ≥1:80 to 1:100 Hemagglutination Rapid in-house serologic assays have also been used to make
inhibition (HI) decisions regarding isolation or euthanasia in shelter situations,
Rabies ≥0.5 IU/mL Fluorescent antibody through identification of immune animals.28 Unfortunately, it is
virus neutralization not always be possible to know if positive titers represent recent
(FAN) infection, and animals that test positive may still shed virus
and pose a risk to other animals. In young puppies and kittens,
positive results may represent persistent MDA or the presence
specimen, and there is a lack of validated sensitivity and speci- of active immunity, and MDA does not have the same ability
ficity for these assays. Sometimes use of these assays increases to protect against infection. In-house serologic assays can also
costs significantly and delays immunization. In-practice assays be used to decide whether pregnant animals are susceptible in
also are available for detection of antibody responses, and these a shelter environment and thus minimize adverse reactions to
have the potential to overcome problems associated with labo- attenuated live vaccines in this group of animals (see Appendix
ratory quality control and delays in immunization. In-practice I). A study that evaluated the performance of one ELISA assay
assays are generally not quantitative. Although high antibody (Synbiotics TiterCHEK CPV/CPV) found that for CPV antibod-
titers are generally associated with greater protection, an ani- ies, the sensitivity and specificity of the assay was 92% and 94%,
mal with no titer may still be resistant to challenge because respectively, when compared with hemagglutination inhibition;
of CMI, which is not measured. Conversely, an animal with a and for CDV was 76% and 92%, respectively, when compared
titer that is generally regarded as protective has the potential with serum neutralization.29
124 SECTION 3  Basic Principles for Infection Control

vaccine-associated adverse effects that were listed as vaccine


reactions, allergic reactions, anaphylaxis, urticaria, and/or car-
diac arrest were documented within 3 days of vaccine adminis-
tration in 38.2 per 10,000 dogs and 47.4 per 10,000 cats.20,31
Reactions coded as “allergic” or “anaphylaxis” were reported
in approximately 1 in 785 dogs and 1 in 1200 cats. Reactions
coded as anaphylaxis constituted only 5% of these reactions.
Death occurred in 1 in 400,000 dogs and 1 in 125,000 cats that
received vaccines, and all 3 dogs and 1 of the 4 cats that died
received four or more doses of a vaccine (i.e., more than one
vaccine product administrated simultaneously). Most reactions
in dogs (73%) occurred on the day the vaccine was administered
(day 0), 19% occurred on day 1, 6% on day 2, and 3% on
day 3. Data from the UK Veterinary Products Committee report
FIGURE 12-3  Facial edema and hyperemia in a 4-month old intact male Chihuahua indicated anaphylaxis in 1 in 385,000 vaccinated dogs and 1 in
mix after vaccination with a bacterin vaccine. (Courtesy Dr. Stephen D. White, University of 555,000 cats.23
California, Davis Veterinary Dermatology Service.) Vaccines that contain large amounts of adjuvant, certain pre-
servatives, or inactivated bacteria with proinflammatory outer
surface components are more likely to cause reactions. Proteins
Adverse Reactions to Vaccines present in fetal calf serum and stabilizers such as gelatin within
the vaccine may also be responsible for allergic reactions.32
A vaccine is needed if an infectious disease causes significant In the Banfield study, the risk of reactions increased with the
morbidity and mortality. Vaccines should prevent more disease number of vaccine doses (i.e., volume of vaccine in milliliters)
than they cause. In order to produce protective immunity, a vac- administered per office visit.20 Small-breed dogs, such as minia-
cine must stimulate a reaction in an animal, both at the site of ture dachshunds, pugs, Boston terriers, miniature pinschers, and
injection and systemically. This may cause clinical signs. Ide- Chihuahuas, were more susceptible to development of acute vac-
ally, the signs are mild and either unnoticeable or acceptable to cine reactions, and the risk of a vaccine-related adverse increased
the pet owner. In rare situations, adverse reactions are severe as body weight decreased. The risk of vaccine-related adverse
enough to threaten a pet’s life. Sometimes, enhanced efficacy events was 4 times greater in dogs that weighed 5 kg or less than
leads to a reduction in vaccine safety. Veterinarians are encour- in those that weighed more than 45 kg (Figure 12-4). Adverse
aged to report adverse reactions to vaccines to a technical service events increased in frequency with age up until 2 years of age in
veterinarian employed for this purpose by the vaccine manufac- dogs and 1 year of age in cats, after which the frequency progres-
turer. In some countries, the drug company then reports details sively declined to rates lower than that observed in animals less
of the adverse reaction to drug regulatory authorities. An under- than 1 year of age (see Figure 12-4). The decrease in frequency
standing of the true nature and incidence of adverse effects with older age may have occurred because of owners’ unwilling-
associated with vaccination has been hampered by underreport- ness to have their pets vaccinated if a previous reaction occurred.
ing and variable delays between vaccination and the inconsis- Sexually intact dogs were less likely to develop adverse reac-
tent appearance of potential, more chronic systemic adverse tions than neutered dogs, but the opposite was true for cats.20,31
effects.30 In addition, correlation of adverse reactions to vaccine Female cats were more likely to exhibit reactions than male cats.
administration in young animals may be difficult because of the The treatment of choice for anaphylaxis is epinephrine,
uniform and frequent administration of vaccines to this group. together with other supportive treatments such as intravenous
For example, it has been difficult to prove a connection between fluids and supplemental oxygen if necessary. Antihistamines
vaccination for distemper and development of hypertrophic and corticosteroids can be administered to dogs with less severe
osteopathy in Weimaraner dogs. reactions. Vaccination should be avoided in animals with a his-
tory of severe reactions. Pretreatment with an antihistamine
Immune-Mediated Vaccine Reactions could be considered in animals with a history of mild reactions.
Type I Hypersensitivity Reactions These animals should also be monitored closely in the hospital
Type I hypersensitivity reactions occur when allergens cross- for several hours after vaccine administration. It has been sug-
link IgE molecules that are bound to receptors on mast cells gested that in the future, commercial production of low-dose
and basophils and trigger degranulation. Clinical signs of type vaccines for small-breed dogs might be more appropriate, given
I hypersensitivity responses that occur after vaccine administra- their increased risk of reactions and more marked serologic
tion include facial or periorbital edema, urticaria, cutaneous responses to vaccination.33
hyperemia, generalized pruritus, salivation, hypotensive shock,
tachypnea, vomiting, diarrhea, collapse, and even death (Figure Other Hypersensitivity Reactions, including Autoimmune Disease
12-3). Vomiting and respiratory distress are common in cats. Type II hypersensitivity reactions occur when IgG and IgM bind
These signs generally occur within 24 hours of vaccine admin- to cell surface antigens and fix complement, with target cell lysis
istration; anaphylaxis usually begins within minutes. The esti- or removal of target cells by macrophages within reticuloen-
mated incidence of anaphylaxis after vaccination of dogs and dothelial tissues. Concerns have been raised that vaccination
cats is 1 in 5000 to 1 in 50,000 and depends on the vaccines may predispose certain genetically susceptible individuals to
used. One retrospective study evaluated 1.23 million dogs and immune-mediated cytopenias, although as in human medicine,
nearly 0.5 million cats from more than 300 Banfield hospi- studies of dogs and cats to date have failed to conclusively docu-
tals in the United States in 2002 through 2005. In this study, ment vaccines as causes of these and other chronic diseases.21,34
CHAPTER 12  Immunization 125

Immune-mediated hemolytic anemia was suspected to occur


following parvovirus vaccination, possibly due to the hemag-
glutinating properties of the virus and the high antigen mass
in some of these vaccines,35 but a later retrospective case-­
control study found no association.36 Transient thrombocyto-
penia can occur after vaccination in some dogs.37 In one study,
dogs developed antithyroglobulin antibodies after vaccination,
although this was not associated with development of hypothy-
roidism.38 Cats that are vaccinated with Crandell-Rees feline
kidney (CRFK) cell-derived vaccines develop antibodies against
the CRFK proteins alpha-enolase and annexin A2.39 Whether
production of these antibodies has clinical significance remains
to be determined.
Type III hypersensitivity reactions are characterized by
immune-complex deposition in tissues and may be a consequence
of immunization with certain vaccines. For example, anterior
A uveitis and subclinical nephritis developed in 0.4% of dogs
receiving the canine adenovirus-1 (CAV-1) vaccine. This vaccine
has now been replaced by CAV-2 vaccines, which rarely produce
these lesions. A cutaneous vasculitis has been described after
vaccination of dogs and cats with rabies virus vaccines (“rabies
vaccine-induced vasculitis”)40-42 (Figure 12-5). This can occur at
the site of vaccine administration, and in some animals, a mul-
tifocal ischemic dermatopathy and myopathy that affects sites
such as the pinnal margins, periocular areas, tail tip, and paw
pads has been reported to occur 1 to 5 months after the appear-
ance of the initial skin lesion.40 The multifocal dermatopathy
and myopathy has been reported to resolve after treatment with
pentoxifylline and vitamin E. Additional evidence is required
to strengthen the association between the multifocal condition
and rabies vaccination. Similarly, concerns have been raised
about a possible temporal association between vaccination and
B immune-mediated polyarthritis in some dogs,43 but this associa-
FIGURE 12-4  Correlation between (A) body weight and (B) age and the occurrence tion remains unproven. Immune-mediated polyradiculoneuritis,
of vaccine-associated adverse effects within 3 days of vaccination in dogs. (From Moore GE, a type IV hypersensitivity reaction (delayed-type hypersensitiv-
Guptill LF, Ward MP, et al. Adverse events diagnosed within three days of vaccine adminis- ity), occurred after vaccination of dogs with suckling-mouse
tration in dogs. J Am Vet Med Assoc 2005;227(7):1102-1108.) brain-derived inactivated rabies vaccines, which are no longer

A B
FIGURE 12-5  Ischemic dermatopathy suspected to be associated with rabies virus vaccination that involved the pinnae (A) and footpads (B) of a 1-year-old male dachshund.
The rabies vaccine was administered several months before the onset of signs. (Courtesy Dr. Stephen D. White, University of California, Davis Veterinary Dermatology Service.)
126 SECTION 3  Basic Principles for Infection Control

available.44 Subsequent reexposure resulted in more severe and


prolonged paralysis. Granuloma formation at the site of vaccine
administration also represents a type IV hypersensitivity reaction.
Despite the lack of conclusive evidence for an association
between vaccination with currently available vaccines and auto-
immune disease, it is possible that vaccination may be associated
with dysregulation of the immune response in predisposed indi-
viduals. Therefore, vaccination is often withheld if not absolutely
necessary in dogs and cats with a history of autoimmune disease.
Serum titers could also be assessed to gauge the need for specific
immunization in these animals.

Vaccine Organism-Induced Disease


Disease occasionally results from replication of microorganisms
present in a vaccine, although severe disease is uncommon with
currently available vaccines. Fever and lethargy are the most
common adverse effects of vaccination and result from cytokine
production in response to the vaccine. These are transient and
usually resolve within 1 to 2 days. FIGURE 12-6  Two-year-old female toy poodle/maltese terrier mix that developed
In the past, the use of attenuated rabies virus vaccines resulted focal alopecia at the site of vaccination followed by regrowth of hair with an altered color
in ascending paralytic disease in a proportion of cats and dogs. and texture. (Courtesy Nicole Pierce, University of ­California, Davis, Class of 2013.)
This led to a change to inactivated, adjuvanted rabies vaccines.
Administration of attenuated parvovirus vaccines to pregnant for 5 to 7 days. Dogs that develop hepatic necrosis may need
cats and dogs can lead to cerebellar disease in the fetus, and more aggressive supportive care.
these vaccines have the potential to cause severe disease if shed
vaccine virus infects colostrum-deprived neonates that are less Local Cutaneous Reactions and Injection-Site
than 2 weeks of age.2 Some CDV vaccines have been associated Sarcomas
with postvaccinal distemper in young puppies.45 As a result, Local cutaneous reactions are common adverse effects of vac-
vaccination with live attenuated CDV and CPV (dogs) and cination, especially in cats, and include pain, swelling, irritation,
feline panleukopenia virus (cats) vaccines should be avoided in and abscess formation. In dogs, focal alopecia or discoloration
pregnant animals, puppies and kittens less than 6 weeks of age, of the haircoat at the vaccination site can also occur (Figure
and animals receiving potent anticancer chemotherapy drugs. 12-6). Inactivated, adjuvanted vaccines have been most com-
Some CDV vaccine strains, such as Rockborn-like strains, are monly incriminated. Focal cutaneous granulomas and some-
more virulent than others, and these may continue to circulate times permanent focal alopecia have been most commonly
and contribute to distemper in the dog population.46 The safest reported after inactivated rabies vaccine administration to
attenuated live CDV vaccines contain the Onderstepoort strain. breeds such as Maltese terriers and bichon frisé.
Vaccination of certain exotic pet, zoo, and wild animal species, In the late 1980s, an increase in inflammatory injection-site
such as ferrets, with any attenuated live CDV vaccine for dogs reactions at the site of rabies vaccine administration were noted
can also lead to postvaccinal distemper.47,48 For animals with in canine and feline biopsy specimens sent to the University
chronic immunocompromise, the use of inactivated vaccines has of Pennsylvania. Shortly thereafter, sarcomas were observed
been recommended if immunization is deemed necessary. How- at these sites in cats in the United States, with a 25% increase
ever, inactivated vaccines may have reduced efficacy in immuno- each year from 1987 to 1991 (Figure 12-7).52 This followed
compromised animals compared with healthy animals. (1) the change from attenuated live to inactivated rabies vac-
The use of mucosal (e.g., intranasal) vaccines for respiratory cines, (2) increased use of rabies vaccines in cats, and (3) the
pathogens in dogs and cats can be followed by development of introduction of FeLV vaccines. The national incidence of sar-
mild to moderate, transient upper respiratory tract signs. There coma formation is estimated to be 0.6 to 2 sarcomas per 10,000
have been concerns that mucosal Bordetella bronchiseptica cats that are vaccinated.53,54 In contrast, in the United King-
vaccines may cause respiratory disease in immunosuppressed dom, it was 0.21 per 10,000 vaccine doses sold between 1995
humans who inhale the vaccine directly during administration and 1999.22 The interval between tumor development and the
or who contact vaccine organisms that are subsequently shed last rabies vaccine typically ranges from 3 months to 3.5 years.
by the vaccinated dog,49,50 but definitive proof of this is still Most tumors are fibrosarcomas, but other types of sarcomas can
required. Inadvertent parenteral administration of the aviru- also occur. Although development of injection-site sarcomas is
lent live intranasal B. bronchiseptica vaccine to dogs can lead clearly linked to administration of FeLV, rabies, and other vac-
to local injection-site reactions and, occasionally, fatal hepatic cines, development of sarcomas is not related to the use of spe-
necrosis.51 Inadvertent parenteral administration of mucosal cific brands or types of vaccine within an antigen class (with the
B. bronchiseptica vaccines should be treated with subcutane- possible exception of recombinant vaccines56), reuse of syringes,
ous fluids at the site of administration and treatment with an needle gauge, use and shaking of multidose vials, or concomi-
oral antibiotic likely to be effective against B. bronchiseptica, tant viral infection.55 There is also no evidence that aluminum-
such as doxycycline. The ASPCA Poison Control Center also containing vaccines are associated with a higher risk of sarcoma
recommends injection of a gentamicin sulfate solution into development than aluminum-free vaccines, but there has been
the affected area (2 to 4 mg/kg gentamicin sulfate in 10 to concern that adjuvant stimulates an inflammatory response that
30 mL of saline).4 Doxycycline treatment could be continued predisposes to sarcoma formation. A recent study showed that
CHAPTER 12  Immunization 127

A B
FIGURE 12-7  A, Computed tomographic image of an injection-site sarcoma over the scapula of a 12-year old male neutered domestic shorthair cat. B, Discoloration of the haircoat
after radiation therapy.

inactivated vaccines were approximately 10 times more likely to can pool in this region and contribute to a chronic inflamma-
be associated with injection-site sarcomas when administered at tory response. In addition, both groups recommended that the
the pelvic limb site than nonadjuvanted recombinant vaccines.56 sites of administration and the product and batch number be
However, neither attenuated live, recombinant, nor inactivated documented to facilitate the reporting of adverse events. Exces-
vaccines are risk-free, and injection of certain long-acting inject- sive administration of vaccines to cats should be minimized,
able medications (especially glucocorticoids) can also be associ- and alternative routes, such as intranasal immunization, should
ated with sarcoma formation in cats years later.56,57 There is be considered. Owners should be advised to monitor injection
some evidence that administration of cold vaccine may also be sites for 3 months after vaccines are administered. If a lump
more likely to be associated with sarcoma formation, but this forms and increases in size 1 month after vaccination, or persists
requires verification.55 It is currently hypothesized that an indi- beyond 3 months, the owner should have the mass evaluated by
vidual cat’s genetically programmed wound healing response is the veterinarian.
responsible for the development of injection-site sarcomas. In accordance with the VAFSTF recommendations, the ana-
Treatment of injection-site sarcomas involves aggressive sur- tomic location, shape, and size of masses that develop at injec-
gical resection followed by full-course post-operative radiation tion sites should be documented, and an incisional or tru-cut
therapy, because of the high incidence of recurrence. Possible biopsy performed. Fine-needle aspiration is not recommended.
adverse effects of radiation include mild to moderate cutaneous If the mass is malignant, routine laboratory tests and thoracic
burns, hypopigmentation of the hair in the field (see Figure 12-7, radiography should be performed, together with computed
B), and damage to the spinal cord, lungs, and kidneys within the tomography or magnetic resonance imaging of the mass if client
field, although the last is rare. In one study, the median sur- finances allow. A veterinary oncologist should be consulted, and
vival time for cats treated with postsurgical curative radiation if possible, referral to a specialist surgeon or oncologist is rec-
therapy was 43 months.57 Most of these cats had clean margins ommended. Wide excision that includes at least a 2-cm margin
after surgical resection of the tumor. In contrast, the median is necessary, and the entire piece of tissue excised should be sub-
survival times for cats treated with coarse fractionated radiation mitted for histopathology and evaluation of surgical margins.
therapy was 24 months. These cats generally had macroscopic Additional treatment, such as chemotherapy and radiation ther-
disease or dirty margins. Adjuvant chemotherapy with carbo- apy, is often necessary. Cats should be reevaluated at 3-month
platin or single-agent doxorubicin also has been associated with intervals for a year after surgery.
improved outcome.57
In order to prevent death from sarcoma formation in cats, Interference with Diagnostic Test Results
the Vaccine-Associated Feline Sarcoma Task Force (VAFSTF) in Vaccination has the potential to interfere with the results of
North America recommended that rabies vaccines be adminis- assays that detect the antibody response to infection or assays
tered as distally as possible on the right pelvic limb, and leuke- that detect components of a pathogen itself.
mia vaccines be administered as distally as possible on the left Interference with antibody test results can be specific or non-
pelvic limb (rabies right, leukemia left). Care should be taken specific. Nonspecific interference is rarely identified, but results
not to administer the vaccine too proximally or in the flank from cross-reactivity between antibodies to vaccine components
region, because tumors in these locations cannot be resected as (such as albumin) and the reagents used in serodiagnostic tests.
effectively.58 Other core vaccines should be administered over More commonly, specific interference with serodiagnostic tests
the right shoulder. These recommendations were not adopted by for the infection that is targeted by the vaccine occurs. This espe-
the WSAVA, which suggested that the skin of the lateral thorax cially problematic if (1) vaccination does not completely protect
and abdomen be used for vaccination, and vaccination sites be against infection, (2) the results of serologic tests are required
rotated from year to year.2 Both groups recommended that the for diagnosis, and (3) infection is chronic and persistent, and so
interscapular region be avoided, because vaccine constituents identification of recent natural infection through seroconversion
128 SECTION 3  Basic Principles for Infection Control

is usually not possible. For example, the inactivated FIV vaccine ahead of time is not possible. Canine core vaccines include
does not provide 100% protection, but vaccinated cats develop vaccines for CPV, CDV, CAV, and rabies for countries where
antibodies to the vaccine virus. These antibodies are detected by rabies is endemic. The core feline vaccines are those for feline
ELISA and Western blot immunoassays used for diagnosis of ­herpesvirus-1 (FHV-1), feline calicivirus (FCV), FPV, and rabies.
FIV infection. In the absence of a history of vaccination, posi- Noncore vaccines are optional vaccines that should be con-
tive antibody test results indicate active infection.59 PCR can be sidered in light of exposure risk, that is, based on geographic
used to detect FIV infection in infected cats that have a history distribution and the lifestyle of the pet. Vaccines considered
of vaccination with FIV vaccines, but PCR can occasionally be as noncore vaccines for dogs are canine parainfluenza virus,
negative in cats with active infection, so a negative PCR result canine influenza virus, B. bronchiseptica, Leptospira spp.,
does not rule out natural infection with FIV. Vaccine interfer- and Borrelia burgdorferi. Optional or noncore vaccines for
ence with serodiagnosis can occur after vaccination of dogs for cats include FeLV, FIV, virulent FCV, Chlamydia felis, and
influenza or leptospirosis, but because both of these diseases B. bronchiseptica vaccines.
are acute, seroconversion can be used for diagnosis of recent Several other vaccines are currently available on the mar-
infection in vaccinated dogs. Some serologic assays differentiate ket. For dogs, these are vaccines for canine coronavirus, CAV-
between vaccinated and naturally infected animals (DIVA). For 1, and rattlesnake envenomation. The reports of the American
example, serologic assays that detect the C6 antigen of Borrelia Veterinary Medical Association (AVMA) and the AAHA canine
burgdorferi do not detect antibodies that result from immuni- vaccine task force have listed the first two vaccines as not gen-
zation with Lyme vaccines. The development of recombinant erally recommended, because “the diseases are either of little
vaccines that stimulate a pattern of antibody responses that dif- clinical significance or respond readily to treatment,” evidence
fer from those that result from natural infection can help to for efficacy of these vaccines is minimal, and they may “produce
overcome issues related to differentiation of naturally infected adverse events with limited benefit.” Currently, information
and vaccinated animals. regarding the efficacy of the canine rattlesnake vaccine is insuf-
Interference with the results of assays that detect the patho- ficient. For cats, the feline infectious peritonitis (FIP) vaccine is
gen itself (as opposed to the antibody response) occurs after not generally recommended by the AAFP.
vaccination with attenuated live vaccines that are shed by ani-
mals after vaccination. For example, cats may test positive using SUGGESTED READINGS
ELISA assays for FPV antigen after vaccination with attenuated Baker C, Pickering L, Chilton L, et al. General recommendations on immu-
live FPV vaccines, although for some assays, the rate at which nization—recommendations of the Advisory Committee on Immuniza-
this occurs is very low.60 PCR tests can be positive for extended tion Practices (ACP). MMWR Recomm Rep. 2011;60(2):1-64.
periods after vaccination with attenuated live vaccines. In some Day MJ, Horzinek MC, Schultz RD. Guidelines for the vaccination of
cases, sequence analysis of the PCR product can sometimes dogs and cats. Compiled by the Vaccination Guidelines Group of the
World Small Animal Veterinary Association. J Small Anim Pract.
allow differentiation between vaccine and field strains, but
2007;48(9):528-541.
this is currently performed only on a research basis.61 Rapid Larson LJ, Newbury S, Schultz RD. Canine and feline vaccinations and
PCR assays have also been designed that differentiate between immunology. In: Miller L, Hurley K, eds. Infectious Disease Manage-
vaccine and field strains of some pathogens, such as CDV or ment in Animal Shelters. Ames, IA: Wiley-Blackwell; 2009:61-82.
B. bronchiseptica.62,63 Quantification of organism numbers Moore GE, HogenEsch H. Adverse vaccinal events in dogs and cats. Vet
present in a specimen through the use of real-time PCR assays Clin North Am Small Anim Pract. 2010;40:393-407.
(e.g., for CDV) may shed light on whether natural infection Paul MA, Appel M, Barrett R, et  al. Report of the American Animal
(high organism load) or vaccination (low organism load) has Hospital Association (AAHA) Canine Vaccine Task Force: executive
occurred. summary and 2003 canine vaccine guidelines and recommendations.
J Am Anim Hosp Assoc. 2003;39(2):119-131.
The 2006 American Association of Feline Practitioners Feline Vaccine
Vaccine Selection Advisory Panel Report. J Am Vet Med Assoc 229:1405–1441 (also
http://www.aafponline.org/resources/practice_guidelines.htm).
The advantages and disadvantages of vaccines and vaccine
combinations that are currently available on the market are
provided in the relevant sections of this book for each infec-
REFERENCES
tion entitled “Immunity and Vaccination.” Suggested vaccina- 1. Delves PJ, Martin SJ, Burton DR, et al. eds. Roitt’s Essential Immu-
tion schedules for individual pets and shelter animals that are nology. 12th ed. Ames, IA: Wiley-Blackwell; 2011.
based on recommendations provided by the American Animal 2. Day MJ, Horzinek MC, Schultz RD. WSAVA guidelines for the
vaccination of dogs and cats. J Small Anim Pract. 2010;51(6):1-32.
Hospital Association (AAHA), the American Association for
3. Larson LJ, Newbury S, Schultz RD. Canine and feline vaccina-
Feline Practitioners (AAFP), the European Society for Feline tions and immunology. In: Miller L, Hurley K, eds. Infectious Dis-
Medicine (ESFM), and WSAVA are summarized in tables in ease Management in Animal Shelters. Ames, IA: Wiley-Blackwell;
Appendix.2,15,64-73 2009:61-82.
To facilitate vaccine selection, vaccines for dogs and cats have 4. De Gregorio E, D’Oro U, Wack A. Immunology of TLR-­
been divided by various task forces into core vaccines, noncore independent adjuvants. Curr Opin Immunol. 2009;21(3):339-345.
vaccines, and those that are generally not recommended. 5. Cox JC, Coutler AR. Adjuvants—a classification and review of
Core vaccines are recommended for all animals with an their modes of action. Vaccine. 1997;15(3):248-256.
unknown vaccination history. The diseases involved have sig- 6. Abdelmagid OY, Larson L, Payne L, et al. Evaluation of the efficacy
nificant morbidity and mortality and are widely distributed, and and duration of immunity of a canine combination vaccine against
virulent parvovirus, infectious canine hepatitis virus, and distemper
in general, vaccination results in good protection from disease.
virus experimental challenges. Vet Ther. 2004;5(3):173-186.
All shelter animals should be vaccinated with core vaccines 7. Schultz RD. Duration of immunity for canine and feline vaccines: a
before entry to a shelter or at the time of entry if immunization review. Vet Microbiol. 2006;117(1):75-79.
CHAPTER 12  Immunization 129

8. Schultz RD, Thiel B, Mukhtar E, et al. Age and long-term protec- 31. Moore GE, DeSantis-Kerr AC, Guptill LF, et  al. Adverse events
tive immunity in dogs and cats. J Comp Pathol. 2010;142(Suppl 1): after vaccine administration in cats: 2,560 cases (2002-2005). J Am
S102-S108. Vet Med Assoc. 2007;231:94-100.
9. Larson LJ, Schultz RD. Effect of vaccination with recombinant 32. Ohmori K, Masuda K, Maeda S, et  al. IgE reactivity to vac-
canine distemper virus vaccine immediately before exposure under cine components in dogs that developed immediate-type aller-
shelter-like conditions. Vet Ther. 2006;7(2):113-118. gic reactions after vaccination. Vet Immunol Immunopathol.
10. Brun A, Chappuis G, Précausta P, et  al. Immunisation against 2005;104(3-4):249-256.
panleukopenia: early development of immunity. Comp Immunol 33. Day MJ. Vaccine safety in the neonatal period. J Comp Pathol.
Microbiol Infect Dis. 1979;1(4):335-339. 2007;137:S51-S56.
11. Phillips TR, Jensen JL, Rubino MJ, et al. Effects of vaccines on the 34. Edwards DS, Henley WE, Ely ER, et al. Vaccination and ill-health
canine immune system. Can J Vet Res. 1989;53(2):154-160. in dogs: a lack of temporal association and evidence of equivalence.
12. Mastro JM, Axthelm M, Mathes LE, et  al. Repeated suppres- Vaccine. 2004;22(25-26):3270-3273.
sion of lymphocyte blastogenesis following vaccinations of CPV- 35. Duval D, Giger U. Vaccine-associated immune-mediated hemolytic
immune dogs with modified-live CPV vaccines. Vet Microbiol. anemia in the dog. J Vet Intern Med. 1996;10(5):290-295.
1986;12(3):201-211. 36. Carr AP, Panciera DL, Kidd L. Prognostic factors for mortal-
13. Strasser A, May B, Teltscher A, et al. Immune modulation follow- ity and thromboembolism in canine immune-mediated hemo-
ing immunization with polyvalent vaccines in dogs. Vet Immunol lytic anemia: a retrospective study of 72 dogs. J Vet Intern Med.
Immunopathol. 2003;94(3-4):113-121. 2002;16(5):290-295.
14. Foley JE, Orgad U, Hirsh DC, et al. Outbreak of fatal salmonellosis 37. Straw B. Decrease in platelet count after vaccination with dis-
in cats following use of a high-titer modified-live panleukopenia temper-hepatitis (DH) vaccine. Vet Med Small Anim Clin.
virus vaccine. J Am Vet Med Assoc. 1999;214(1):67-70:43-44. 1978;73(6):725-726.
15. Welborn LV, DeVries JG, Ford R, et al. 2011 AAHA canine vac- 38. Scott-Moncrieff JC, Azcona-Olivera J, Glickman NW, et al. Evalu-
cination guidelines. J Am Anim Hosp Assoc. 2011;47:1-42. ation of antithyroglobulin antibodies after routine vaccination in
16. Baker C, Pickering L, Chilton L, et al. General recommendations pet and research dogs. J Am Vet Med Assoc. 2002;221:515-521.
on immunization—recommendations of the Advisory Commit- 39. Whittemore JS, Hawley JR, Jensen WA, et al. Antibodies against
tee on Immunization Practices (ACP). MMWR Recomm Rep. Crandell Rees feline kidney (CRFK) cell line antigens, alpha-­
2011;60(2):1-64. enolase, and annexin A2 in vaccinated and CRFK hyperinoculated
17. Scott FW, Geissinger CM. Long-term immunity in cats vac- cats. J Vet Intern Med. 2010;24(2):306-313.
cinated with an inactivated trivalent vaccine. Am J Vet Res. 40. Vitale CB, Gross TL, Magro CM. Vaccine-induced ischemic der-
1999;60(5):652-658. matopathy in the dog. Vet Dermatol. 1999;10:131-142.
18. King GE, Hadler SC. Simultaneous administration of childhood 41. Wilcock BP, Yager JA. Focal cutaneous vasculitis and alope-
vaccines: an important public health policy that is safe and effica- cia at sites of rabies vaccination in dogs. J Am Vet Med Assoc.
cious. Pediatr Infect Dis. 1994;13(5):394-407. 1986;188(10):1174-1177.
19. Offit PA, Quarles J, Gerber MA, et  al. Addressing parents’ con- 42. Nichols PR, Morris DO, Beale KM. A retrospective study of
cerns: do multiple vaccines overwhelm or weaken the infant’s canine and feline cutaneous vasculitis. Vet Dermatol. 2001;12(5):
immune system? Pediatrics. 2002;109(1):124-129. 255-264.
20. Moore GE, Guptill LF, Ward MP, et al. Adverse events diagnosed 43. Kohn SL, Garner M, Bennett D, et al. Polyarthritis following vacci-
within three days of vaccine administration in dogs. J Am Vet Med nation in four dogs. Vet Comp Orthoped Traumatol. 2003;16:6-10.
Assoc. 2005;227(7):1102-1108. 44. Gehring R, Eggars B. Suspected post-vaccinal acute polyradiculo-
21. Orenstein WA, Pickering LK, Mawle A, et  al. Immunization. neuritis in a puppy. J S Afr Vet Assoc. 2001;72(2):96.
In: Mandell GL, Bennett JE, Dolin R, eds. Principles and Prac- 45. Cornwell HJ, Thompson H, McCandlish IA, et al. Encephalitis in
tice of Infectious Diseases. 7th ed. Philadephia, PA: Elsevier; dogs associated with a batch of canine distemper (Rockborn) vac-
2010:3917-3949. cine. Vet Rec. 1988;122(3):54-59.
22. Coffey TJ, Werling D. Therapeutic targeting of the innate immune 46. Martella V, Blixenkrone-Møller M, Elia G, et al. Lights and shades
system in domestic animals. Cell Tissue Res. 2011;343(1):251-261. on an historical vaccine canine distemper virus, the Rockborn
23. Day MJ. Vaccine adverse effects: fact and fiction. Vet Microbiol. strain. Vaccine. 2011;29(6):1222-1227.
2006;117:51-58. 47. Carpenter JW, Appel MJ, Erickson RC, et al. Fatal vaccine-induced
24. Kennedy LJ, Lunt M, Barnes A, et  al. Factors influencing the canine distemper virus infection in black-footed ferrets. J Am Vet
antibody response of dogs vaccinated against rabies. Vaccine. Med Assoc. 1976;169(9):961-964.
2007;25(51):8500-8507. 48. Bush M, Montali RJ, Brownstein D, et  al. Vaccine-induced
25. Berndtsson LT, Nyman AK, Rivera E, et  al. Factors associated canine distemper in a lesser panda. J Am Vet Med Assoc.
with the success of rabies vaccination of dogs in Sweden. Acta Vet 1976;169(9):959-960.
Scand. 2011;53:22. 49. Gisel JJ, Brumble LM, Johnson MM. Bordetella bronchiseptica
26. Jónsdóttir I. Maturation of mucosal immune responses and influ- pneumonia in a kidney-transplant patient after exposure to recently
ence of maternal antibodies. J Comp Pathol. 2007;137(Suppl 1): vaccinated dogs. Transpl Infect Dis. 2010;21(1):73-76.
S20-S26. 50. Berkelman RL. Human illness associated with the use of veterinary
27. Siegrist CA. The challenges of vaccine responses in early life: vaccines. Clin Infect Dis. 2003;37(3):407-414.
selected examples. J Comp Pathol. 2007;137(Suppl 1):S4-S9. 51. Toshach K, Jackson MW, Dubielzig RR. Hepatocellular necrosis
28. Crawford PC, Levy JK, Leutenegger C. Use of antibody titers and associated with the subcutaneous injection of an intranasal Bor-
quantitative PCR as risk assessment tools for management of an out- detella bronchiseptica–canine parainfluenza vaccine. J Am Anim
break of canine distemper and parvovirus. Denver, CO: Proceedings Hosp Assoc. 1997;33(2):126-128.
of the 2011 American College of Veterinary Internal Medicine; 2011. 52. Hendrick MJ, Shofer FS, Goldschmist MH, et al. Comparison of
29. Litster AL, Pressler B, Volpe A, et al. Accuracy of a point-of-care fibrosarcomas that developed at vaccination sites and nonvaccina-
ELISA test kit for predicting the presence of protective canine par- tion sites in cats: 239 cases (1991-1992). J Am Vet Med Assoc.
vovirus and canine distemper virus antibody concentrations in 1994;205(10):1425-1429.
dogs. Vet J. 2012;193:363-366. 53. Richards JR, Elston TH, Ford RB, et  al. The 2006 American
30. Moore GE, HogenEsch H. Adverse vaccinal events in dogs and Association of Feline Practitioners Feline Vaccine Advisory Panel
cats. Vet Clin North Am Small Anim Pract. 2010;40:393-407. Report. J Am Vet Med Assoc. 2006;229(9):1405-1441.
130 SECTION 3  Basic Principles for Infection Control

54. Gobar GM, Kass PH. World Wide Web-based survey of vaccina- 64. American Animal Hospital Association (AAHA) Canine Vaccine
tion practices, postvaccinal reactions, and vaccine site-associated Taskforce. 2006 AAHA canine vaccine guidelines. J Am Anim
sarcomas in cats. J Am Vet Med Assoc. 2002;220(10):1477-1482. Hosp Assoc. 2006;42(2):80-89.
55. Kass PH, Spangler WL, Hendrick MJ, et  al. Multicenter case- 65. Klingborg DJ, Hustead DR, Curry-Galvin EA, et al. AVMA’s prin-
control study of risk factors associated with development of ciples of vaccination. J Am Vet Med Assoc. 2001;219:575-576:
vaccine-associated sarcomas in cats. J Am Vet Med Assoc. (also http://www.avma.org/policies/vaccination.htm).
2003;223(9):1283-1292. 66. Truyen U, Addie D, Belçk S, et  al. Feline panleukopenia. ABCD
56. Srivastav A, Kass PH, McGill LD, et  al. Comparative vaccine-­ guidelines on prevention and management. J Feline Med Surg.
specific and other injectable-specific risks of injection-site sarcomas 2009;11(7):538-546.
in cats. J Am Vet Med Assoc. 2012;241:595-602. 67. Thiry E, Addie D, Belçk S, et al. Feline herpesvirus infection. ABCD
57. Eckstein C, Guscetti F, Roos M, et al. A retrospective analysis of guidelines on prevention and management. J Feline Med Surg.
radiation therapy for the treatment of feline vaccine-associated sar- 2009;11(7):547-555.
coma. Vet Comp Oncol. 2009;7(1):54-68. 68. Radford AD, Addie D, Belçk S, et  al. Feline calicivirus infection.
58. Shaw SC, Kent MS, Gordon IK, et al. Temporal changes in charac- ABCD guidelines on prevention and management. J Feline Med
teristics of injection-site sarcomas in cats: 392 cases (1990-2006). Surg. 2009;11(7):556-564.
J Am Vet Med Assoc. 2009;234(3):376-380. 69. Lutz H, Addie D, Belçk S, et  al. Feline leukemia. ABCD guide-
59. Levy JK, Crawford PC, Slater MR. Effect of vaccination against lines on prevention and management. J Feline Med Surg.
feline immunodeficiency virus on results of serologic testing in cats. 2009;11(7):565-574.
J Am Vet Med Assoc. 2004;225(10):1558-1561. 70. Frymus T, Addie D, Belçk S, et al. Feline rabies. ABCD guidelines
60. Patterson EV, Reese MJ, Tucker SJ, et  al. Effect of vaccination on prevention and management. J Feline Med Surg. 2009;11(7):
on parvovirus antigen testing in kittens. J Am Vet Med Assoc. 585-593.
2007;230(3):359-363. 71. Addie D, Belçk S, Boucraut-Baralon C, et al. Feline infectious peri-
61. Hirasawa T, Yono K, Mikazuki K. Differentiation of wild- and tonitis. ABCD guidelines on prevention and management. J Feline
vaccine-type canine parvoviruses by PCR and restriction-enzyme Med Surg. 2009;11(7):594-604.
analysis. Zentralbl Veterinarmed B. 1995;42(10):601-610. 72. Gruffydd-Jones T, Addie D, Belçk S, et  al. Chlamydophila felis
62. Si W, Zhou S, Wang Z, et  al. A multiplex reverse transcription- infection. ABCD guidelines on prevention and management. J Feline
nested polymerase chain reaction for detection and differentiation Med Surg. 2009;11(7):605-609.
of wild-type and vaccine strains of canine distemper virus. Virol J. 73. Egberink H, Addie D, Belçk S, et  al. Bordetella bronchiseptica
2010;7:86. infection in cats. ABCD guidelines on prevention and management.
63. Iemura R, Tsukatani R, Micallef MJ, et al. Simultaneous analysis of J Feline Med Surg. 2009;11(7):610-614.
the nasal shedding kinetics of field and vaccine strains of Bordetella
bronchiseptica. Vet Rec. 2009;165(25):747-751.
SECTION 1
Viral Diseases

CHAPTER 13

Rabies
Jane E. Sykes and Bruno B. Chomel

and frenzy) (Figure 13-1). The virus is fragile in the environ-


Overview of Rabies ment, and readily inactivated by a variety of disinfectants,
First Described: Circa the second millennium bc, Mesopotamia1 soaps, ultraviolet light, and heat. It can survive up to 3 to 4 days
in carcasses at 20°C, and longer with refrigeration.2,3 Freezing
Cause: Rabies virus (Family Rhabdoviridae, Genus Lyssavirus)
tissues at temperatures less than −20°C may prolong survival of
Primary Mode of Transmission: biting, with inoculation of the virus for years.
saliva containing the virus There are seven lyssavirus genotypes (Table 13-1). Classi-
Affected Hosts: All warm-blooded animals. Highly suscepti- cal rabies virus belongs to genotype 1. The remaining six lys-
ble hosts include wolves, foxes, coyotes, jackals, dogs, cat- savirus genotypes primarily infect bats and less frequently cause
tle, raccoons, skunks, bats, and mongooses. Moderately fatal human encephalitis, which is clinically indistinguishable
susceptible hosts include cats, ferrets, primates, sheep, from classical rabies.4 Thus, although Taiwan, Australia, New
goats, and horses. Zealand, Iceland, the United Kingdom, Japan, most of west-
ern Europe, Fiji, Hawaii, and Guam are designated terrestrial
Geographic Distribution: Classic (genotype 1) rabies is found
rabies-free, lyssaviruses capable of causing fatal encephalitis
worldwide except for Taiwan, Australia, New Zealand, Ice-
in humans exist in some of these regions, including Australia,
land, the United Kingdom, Japan, most of western Europe,
Western Europe, and the United Kingdom (Figure 13-2). The
Fiji, Hawaii, and Guam.
remaining information in this chapter pertains only to genotype
Major Clinical Signs: Fever, lethargy, behavioral changes, 1 rabies virus infections.
pupillary dilation, ataxia, lower motor neuron paresis or Infection with rabies virus most commonly occurs as a result
paralysis (including of the muscles of mastication), ves- of inoculation of virus-containing saliva into a bite wound, in
tibular signs (including a head tilt), dysphagia, dysphonia, the large majority of the cases after a dog bite. Other routes
tremors, seizures, ptyalism. of transmission reported in species other than dogs and cats
Differential Diagnoses: Distemper virus infection; West Nile
virus infection; equine encephalitis virus infection; bacte- Envelope (membrane)
rial, protozoal, or fungal meningoencephalitis; toxins such Glycoprotein
Matrix protein
as strychnine; central nervous system trauma; spinal neo-
plasia; granulomatous meningoencephalitis.
Public Health Significance: Rabies is a deadly zoonosis and
dogs are a major natural reservoir of the virus. In the United
States, most human rabies is associated with bat contact.

Etiologic Agent and Epidemiology


Rabies is a deadly, zoonotic, neurologic disease caused by a RNA genome
bullet-shaped, enveloped RNA virus that belongs to the genus helical coil
Lyssavirus (from Lyssa, the Greek goddess of madness, rage, FIGURE 13-1  Structure of a rabies virus virion.

132
CHAPTER 13  Rabies 133

TABLE 13-1
Lyssaviruses Belonging to the Family Rhabdoviridae
Genotype Virus Source Distribution
1 Rabies virus Dog, fox, raccoon, bat, others Widespread
2 Lagos bat virus Bats, cats, not in human beings Africa (rare)
3 Mokola Shrews, cats, dog Africa
4 Duvenhage Insectivorous bats Africa (rare)
5 and 6 European bat lyssavirus Insectivorous bats Netherlands, United Kingdom, Denmark,
types 1 and 2 Germany, Spain, France, Poland, Hungary,
Russian Federation, Ukraine, Switzerland
7 Australian bat lyssavirus Flying fox (fruit bats); insectivorous bats Australia; Philippines (serologic evidence)

FIGURE 13-2  Global distribution of lyssaviruses. Distribution of rabies virus (red). Areas where rabies-related viruses only, and not classic rabies genotype 1, are documented are clas-
sified by the WHO as “rabies free” (dark blue). White areas are free of rabies and rabies-related viruses. (Redrawn from Warrell MJ, Warrell DA. Rabies and other lyssavirus diseases. Lancet
2004;363:959-969.)

are rare and include corneal or solid-organ transplantation in are highly susceptible to infection. Moderately susceptible hosts
human patients5; aerosol transmission, such as that occurring include cats, ferrets, primates, sheep, goats, and horses. Marsu-
within caves containing large numbers of bats6,7; and transmis- pials, including opossums, have a low degree of susceptibility.
sion after ingestion of infected tissues or milk.6,8 Some bird species can be experimentally infected but do not
All warm-blooded animals are susceptible to rabies, although develop clinical signs and are unlikely hosts.9
dogs, wild carnivores, and bats are considered the main natural Not every bite from a rabid animal leads to rabies virus infec-
reservoirs of rabies virus. In general, wolves, foxes, coyotes, tion, and infection may not always culminate in death, unless
jackals, dogs, cattle, raccoons, skunks, bats, and mongooses clinical signs develop. Factors influencing the outcome after a
134 SECTION 1  Viral Diseases

A B
FIGURE 13-3  A, Reported cases of rabies that involve cats and dogs, by county, 2010. Histogram represents numbers of counties in each category for total number of cats and dogs
submitted for testing. Note the overlap with the distribution of wildlife reservoir hosts in B. B, Distribution of major rabies virus variants among mesocarnivore reservoirs in the United
States. (A and B modified from Blanton JD, Palmer D, Dyer J, et al. Rabies surveillance in the United States during 2010. J Am Vet Med Assoc 2011;239:773-783.)

bite from a rabid animal include the proximity of the bite site dogs have also been imported into the United States from other
to the central nervous system (CNS), the degree of innervation countries where canine rabies variants are endemic.13,14 In
of the bite site, the age of the host (young animals are more Europe, the major wildlife reservoir species have been foxes
susceptible), the amount of virus inoculated, and the neuroin- and raccoon dogs, and in South Africa, jackals and mongooses
vasiveness of the rabies virus variant involved. If left untreated, predominate. In western Europe, fox rabies has largely been
up to 60% of humans bitten by a rabid dog on the head or neck eliminated as a result of mass vaccination campaigns. Spillover
develop rabies, compared with up to 40% of those bitten on the occurs from wildlife reservoir hosts to domestic animal species,
hand, and 10% of those bitten on the trunk or leg.10 which subsequently contact humans. Nevertheless, the vast
Rabies viruses that circulate within a certain geographic majority of human cases in North America result from contact
region are adapted to specific dominant reservoir hosts (e.g., bat with infected bats, which are present in all states except Hawaii
rabies virus variants or raccoon rabies virus variants). All rabies (see Public Health Aspects). Rabies in rodents and lagomorphs
virus variants can infect and cause disease in animals other than is rare, possibly because they are always killed by the bite of
their reservoir hosts (spillover hosts). In all host species, rabies a rabid animal. In the United States, most reports have been
is usually an acute fatal illness, although some animals, such as from woodchucks in raccoon endemic areas.15 A few cases of
bats and skunks, have a more prolonged course of illness than rabies in pet ferrets, rabbits, and a pet guinea pig have been
others, such as foxes, coyotes, or raccoons. The possibility of a described.16
subclinical carrier state, especially in bats, has been suggested Although rabies occurs in cats and dogs of any age, it is most
based on the detection of rabies virus antibody and, rarely, the frequently reported in young dogs and cats, with a median age
virus itself in some reservoir host species that are not showing of 1 year for both cats and dogs in one study; 75% of animals
signs of illness.4 were 3 years of age or younger.17 Rabies can occur in puppies
Over the past 50 years, owing to mandatory vaccination and and kittens that have not yet reached the age at which routine
control programs, domestic animal rabies in the United States has rabies vaccination is approved, which is an emerging issue as a
decreased in prevalence, whereas wildlife rabies has increased. result of illegal importation of pet dogs from Mexico through
The dog rabies virus variant was considered eradicated from the California and Texas. Male dogs may be slightly more pre-
United States at the beginning of the 21st century. Currently, disposed, and most dogs and cats with rabies have not been
cats are more commonly reported with rabies in the United neutered and live in rural environments, frequently being unre-
States (about 300 cases/year) than dogs (fewer than 100 cases/ strained at night.11,17 In the United States, some degree of sea-
year), most likely reflecting rabies vaccination practices ­(Figure sonality occurs for rabies in dogs and cats, which correlates
13-3, A). No feline rabies virus variant has been described. with seasonal increases in rabid raccoons and skunks.11,17 For
The geographic distribution of feline and canine rabies fol- the majority of cats and dogs with rabies, a history of known
lows that of regions where wildlife rabies is endemic. Most contact with wild animals or a wound is lacking.
cats and dogs are infected with the rabies virus variant that is
associated with the major wildlife reservoir host in their respec- Clinical Features
tive geographic location (see Figure 13-3, B). Rabid cats are
most often reported from the northeastern United States, mid- Signs and Their Pathogenesis
Atlantic, and south-Atlantic states, with lesser numbers from After inoculation into the subcutaneous tissues and muscle,
the north and south central states. The eastern distribution rabies virus replicates locally within muscle cells and then
of feline rabies follows the distribution of raccoon rabies.11,12 attaches to peripheral nerve endings. Local replication around
Rarely, cats are infected from bats. Rabies in dogs and cats is the bite site can continue for months before the virus enters
most commonly reported from the north central, south central, peripheral sensory and motor nerve endings, with the nicotinic
and Atlantic states (see Figure 13-3, A). Dogs may be infected acetylcholine receptor being the main receptor for the virus. The
with rabies virus variants from skunks or raccoons. Infected virus then travels in a retrograde fashion up nerve axons at a
CHAPTER 13  Rabies 135

occur. Unfortunately, rabies is infrequently suspected as the


TABLE 13-2 possible diagnosis at the time rabid animals are examined by
Major Clinical Findings in 183 Cats and 119 Dogs with Rabies veterinarians.11,17
When it occurs, the prodromal phase lasts 2 to 3 days in dogs
in the United States
and 1 to 2 days in cats, and is characterized by a variable fever,
licking or chewing at the bite site, and behavioral changes. Dogs
Sign Percent of Cats Percent of Dogs
and cats may become lethargic, anorexic, apprehensive, restless,
Aggression 55 31 or reclusive, and vomiting may occur.11 Some animals become
Ataxia 30 49 more docile or affectionate. Pupillary dilation, sometimes with
Irritability 34 23 decreased pupillary light reflexes, may occur. The furious
phase occurs in approximately two thirds of affected cats and
Anorexia 22 38 dogs22 and lasts 0 to 7 days. Clinical signs result from forebrain
Lethargy 20 37 involvement and include irritability, anxiousness or excitabil-
Hypersalivation 14 41 ity, hyperesthesia, hypersalivation, vocalization, roaming, and
aggression. Affected animals may try to eat foreign objects,
Dysphagia 10 30
which may become lodged within the gastrointestinal tract, or
Lameness 18 16 they may attack their surroundings or moving objects. Some
Limb paralysis 17 29 animals develop ataxia, vestibular signs, and grand mal sei-
Jaw paralysis 3 29 zures. Tremors; staring, or a wild, spooky, or blank look in the
eyes; increased vocalization; and compulsive running can occur
Dysphonia 16 9
cats, which also often develop aggressive behavior.11,17,23 The
Hyperesthesia 10 17 paralytic phase develops 1 to 10 days after the onset of clinical
Seizures 8 14 signs and is characterized by flaccid paralysis, which is ascend-
Fever 3 14 ing and often initially involves the bitten extremity. Neurologic
examination reveals flaccid paralysis with absent segmental
From Eng TR, Fishbein DB. Epidemiologic factors, clinical findings, reflexes. Laryngeal paralysis may lead to a change in the sound
and vaccination status of rabies in cats and dogs in the United States or pitch of a dog’s bark or cat’s meow. Hypersalivation results
in 1988. National Study Group on Rabies. J Am Vet Med Assoc from paralysis of the pharyngeal muscles, and a “dropped jaw”
1990;197:201-209. can occur as a result of masticatory muscle paralysis, especially
in dogs, which may appear to owners as if they are choking.
rate of 3 mm/hr.6 Once the virus is within the CNS, there is mas- If euthanasia is not performed, coma and death usually
sive viral replication, with cell-to-cell transmission of the virus occurs within a week of the onset of signs, with a few ani-
across synaptic junctions. The spinal cord, medulla oblongata, mals dying as long as 10 days after the onset of illness. Death
periaqueductal gray matter, limbic system, and cerebellum are is associated with multiorgan failure, especially cardiac and
particularly affected. The virus also moves outwards from the respiratory failure. Recovery from rabies is extremely rare.
CNS in somatic and autonomic nerves and is deposited in a vari- Experimentally, some dogs have recovered from clinical rabies
ety of tissues, including cardiac and skeletal muscle, the eye, the days to months after being infected with certain rabies virus
kidney, pancreas, nerves around hair follicles, and the salivary strains.24,25
glands. Production of new virions through budding from the
plasma membranes occurs primarily within the salivary glands, Diagnosis
which results in the shedding of virus that can be transmitted to
other hosts. Thus, the presence of virus in the saliva indicates Rabies should be suspected in dogs and cats evaluated for sud-
that the CNS has been infected. In some animals, death occurs den onset of behavioral changes and flaccid paralysis, especially
before the saliva becomes infected. Virus is shed by some dogs in rabies-endemic regions, or in animals imported from rabies-
for up to 13 days before the onset of clinical signs.18 endemic countries. Although a particularly high degree of suspi-
Clinical signs probably result from impaired neuronal func- cion is warranted for animals that lack a vaccination history for
tion6 and occur after an incubation period that ranges from just rabies, the disease has been reported in partially and completely
over a week to 6 months. Most dogs and cats develop disease vaccinated dogs and cats,11,17,23 so a history of rabies vaccina-
1 to 2 months after exposure. In general, the closer the bite site tion should not be used to dismiss suspicion for rabies.
is to the CNS, the shorter the incubation period. The incuba-
tion period is also influenced by the host species, age, degree of Laboratory Abnormalities
innervation of the bite site, neuroinvasiveness of the rabies virus No specific or characteristic hematological or biochemical
variant, and the amount of virus inoculated. Although 90% of changes have been reported in dogs and cats with rabies. Mild
humans develop disease within 6 months of exposure, incuba- anemia was reported in one dog.26 Analysis of the cerebrospinal
tion periods of 6 years or more have been reported.19-21 fluid (CSF) may be unremarkable or show an increase in CSF
The clinical presentations of rabies virus infection have been protein concentration and mild to marked CSF pleocytosis. Small
divided into excitatory (“furious”) and paralytic (“dumb”) lymphocytes account for most of the differential cell count.26
forms. Three phases have been described in the progression Electromyography in one dog revealed abnormalities con-
of the disease, the prodromal, furious, and paralytic phases, sistent with impaired neuromuscular transmission, including
but the stages are variable and may overlap, and signs may moderate fibrillations, positive sharp waves, and an absent
be atypical (Table 13-2). Infrequently, there is a history of a M wave.26 Changes suggestive of denervation have also been
wound, or wounds are still present at the time neurologic signs described in humans with rabies.27
136 SECTION 1  Viral Diseases

TABLE 13-3
Diagnostic Assays Available for Rabies in Dogs and Cats
Assay Specimen type Target Performance
Direct fluorescent Brain, spinal cord, skin Rabies virus N High sensitivity and specificity on brain tissue; because
antibody stain- biopsies (latter not (nucleocapsid) protein a single negative result does not rule out rabies, con-
ing or immuno- recommended in firmatory tests such as mouse inoculation or reverse-
histochemistry animals), other extra­ transcription polymerase chain reaction (RT-PCR)
cranial tissues are required for specimens testing negative. Sensitiv-
ity when used on extracranial tissues is relatively low.
Serology Serum or cerebrospinal Antirabies virus Virus neutralization assays are the gold standard for
fluid (CSF) antibodies serology, although ELISA assays are also avail-
able. Rarely useful for diagnosis of rabies, as the
antibody response is delayed and so titers are often
negative. Titers often determined before importa-
tion of animals into rabies-free countries.
Histopathology Brain, spinal cord Intracytoplasmic Detection of Negri bodies has low sensitivity.
eosinophilic inclusions
(Negri bodies)
RT-PCR Brain, spinal cord, saliva, Rabies virus RNA Rapid (within hours), has the potential to be very
CSF, extracranial tissues sensitive and specific; can be used to identify rabies
genotypes and strains.

Diagnostic Imaging be performed, and the entire head should be submitted as soon
Magnetic Resonance Imaging Findings as possible to public health authorities, so as not to delay post-
Findings on MRI in dogs with rabies have included hyperintense exposure prophylaxis measures for exposed individuals (see
lesions on T2-weighted images, especially in the midbrain, thal- Public Health Aspects). Opening the skull using a saw generates
amus, and basal ganglia, that do not enhance with contrast.28 aerosols and is not recommended. Although not optimal, if the
In human patients with rabies, no significant abnormalities head is not available, the spinal cord can be submitted. Tissues
may be present on MRI, or it may show hyperintensities on should be refrigerated and not frozen for optimal test sensitiv-
T2-weighted images, especially in association with the gray mat- ity. Approved shipping containers should be used, which must
ter. Lesions are frequently described in the medulla, pons, basal be labeled with appropriate warnings.
ganglia, and thalamus and reflect concentrations of rabies virus Antemortem, DFA testing has been used to detect rabies
antigen.29 Contrast enhancement can be present in comatose virus in corneal touch impressions and frozen skin biopsy sec-
human patients but is usually not detected early in the course tions from the nuchal area in humans33,34 or the region of the
of illness. Computed tomography commonly does not reveal whiskers in dogs and cats. The sensitivity of this method is
lesions in humans with rabies. as low as 25%, and it should not be used for clinical diag-
nosis in dogs and cats suspected to have rabies. DFA stain-
Microbiologic Testing ing has also been used for detection of rabies virus antigen in
Diagnostic assays for rabies in dogs and cats are listed in extracranial tissues at necropsy, including the tongue, salivary
Table 13-3. glands, adrenal glands, pancreas, gastrointestinal tract, and
myocardium.35,36
Direct Fluorescent Antibody Testing and Immunohistochemistry
Rabies virus diagnosis is generally based on direct fluores- Serologic Diagnosis
cent antibody (DFA) staining for virus on impression smears Serologic tests for rabies virus antibodies are often negative in
of brain tissue obtained at necropsy. This is rapid, sensitive, affected animals, as a result of immune evasion by the virus dur-
and economical and is performed only in qualified laboratories ing the incubation period and possibly also virus-induced immu-
that have been approved by local or state public health depart- nosuppression after the onset of clinical signs. If seroconversion
ments. In the United States, a national standardized protocol for occurs, it is typically late in the course of illness. Prior vaccina-
rabies testing is published by the Centers for Disease Control tion for rabies may result in confusing results. Determination
and Prevention.30 False-negative results with DFA staining are of acute and convalescent titers on serum and determination of
rare when compared with mouse inoculation testing,31 and all CSF antibody titers can aid diagnosis of human rabies.34 In dogs
animals with virus in their saliva test positive. A direct rapid and cats, serologic testing is most commonly used to document
immunohistochemistry test is also available, which has a similar proof of prior vaccination, which is required for importation
sensitivity and specificity and does not require a fluorescence into rabies-free countries. It is not suitable for determining the
microscope.32 need for booster vaccinations.37
If rabies is suspected, the animal should be handled with Serologic methods include virus neutralization assays and
extreme caution (see Prevention). Humane euthanasia should ELISA assays.38 The gold standard of rabies serology is the
CHAPTER 13  Rabies 137

rapid fluorescent focus inhibition test (RFFIT), a virus neutral- Treatment and Prognosis
ization assay, which assesses the ability of antibodies to prevent
viral replication in cell culture. The fluorescent antibody virus Immediate euthanasia and testing of dogs and cats suspected to
neutralization (FAVN) test has been accepted as an alternative have rabies is recommended. Once clinical signs have developed,
for determining titers in animals before importation. A mini- rabies is almost always fatal and very few humans have sur-
mum titer of 0.5 U/mL is required before importation to rabies- vived. Most have been children, and several of those individuals
free countries is permitted. had been vaccinated for rabies before the onset of illness. In all
individuals, moderate to severe neurologic sequelae persisted. A
Virus Isolation and Mouse Inoculation person in Texas appeared to survive rabies without the need for
Rabies virus can be isolated in a variety of cell culture lines, intensive care.49 After treatment with induced coma (the “Mil-
although isolation may be less sensitive than nucleic acid-based waukee protocol”), an 8-year old girl survived rabies that may
detection methods.39 Virus isolation and inoculation of mice have been acquired after a scratch from a stray cat.50 Rarely,
with tissue from affected patients can be used to confirm rabies dogs inoculated experimentally with certain canine rabies virus
virus infection by public health authorities when direct fluores- variants have recovered from rabies encephalitis, sometimes with
cent antibody testing of brain tissue is negative. intermittent shedding of rabies virus in saliva after recovery.25

Molecular Diagnosis Using the Polymerase Chain Reaction Immunity and Vaccination
Nucleic acid detection techniques are increasingly employed
to detect rabies virus in tissues and may ultimately replace In most cases of human rabies, no immune response is detect-
techniques such as isolation and histopathology.40 Reverse-­ able at 7 to 10 days after the onset of clinical signs. Rabies virus
transcriptase polymerase chain reaction (RT-PCR) assays may evade immune recognition through inhibition of apoptosis
have been used to rapidly detect rabies virus RNA in dogs and destruction of invading T cells, which may explain the rela-
and humans and, when used on brain tissue, are sensitive and tive lack of an inflammatory response within the CNS.51
specific, the results correlating with those of DFA testing.41,42 Currently available vaccines contain inactivated virus or are
RT-PCR can be used to detect virus in decomposing tissue, canarypox-vectored recombinant rabies (cats) vaccines. Modi-
when the results of other assays might be negative.43,44 Genetic fied live rabies vaccines are no longer available in the United
analysis of resultant PCR products can also be used to iden- States because of their association with occasional postvaccinal
tify lyssavirus genotypes and rabies virus variants (see Table rabies encephalitis in dogs and cats.52 Vaccinia poxvirus-vec-
13-1).45 For antemortem diagnosis in human patients, saliva tored glycoprotein recombinant vaccines are used for wildlife
is more likely to test positive than CSF. A nuchal biopsy speci- vaccination.
men can also be tested.46 The same appears to be true in dogs, All dogs and cats should be vaccinated for rabies at 3 or 4
although again, attempts to make an antemortem diagnosis months of age (depending on state legislation), with a booster
of rabies are not recommended in animals.47 The sensitivity dose 1 year later, then every 3 years with approved inactivated
of RT-PCR for detection of rabies virus in the saliva of rabid vaccines, or annually with recombinant vaccines (see Chapter
dogs was 87%, so a negative result does not rule out a diag- 12). It has been recommended that rabies vaccines be admin-
nosis of rabies. istered as distally as possible in the right pelvic limb, to allow
better understanding of which vaccines are associated with sar-
Pathologic Findings coma formation and to permit complete removal of sarcomas by
Gross Pathologic Findings amputation should they develop.53,54 Canine rabies vaccination
Gross lesions are generally absent in the CNS of dogs and cats is required by law within most of the United States, and vaccines
with rabies. Lesions consistent with a previous wound or trau- must be administered only by (or under the direct supervision
matic event may be present elsewhere on the body. of) a veterinarian.37 Effective vaccination of at least 70% of the
dog population is required to prevent rabies epizootics.55 Own-
Histopathologic Findings ers who fail to comply with state or local requirements should
Rabies virus infection results in a remarkably mild, nonsuppu- be reported to the public health authorities.
rative polioencephalomyelitis. Changes include mild neuronal Although rare, rabies has been reported in vaccinated ani-
degeneration and neuronophagia, which may be followed by mals, especially when vaccinations are not up to date.23 Dogs
neuronal necrosis.26 Lymphoplasmacytic perivascular cuff- and cats less than 1 year of age are not considered immunized
ing and focal microglial hyperplasia are frequently described. until 28 days after the initial vaccination.37 Because of a rapid
The longer the duration of illness, the more pronounced the anamnestic response, an animal is considered fully vaccinated
nonsuppurative inflammatory response. Sometimes, a spongi- immediately after booster vaccination.
form encephalopathy with vacuolization in the gray matter is The use of inactivated rabies virus vaccines in cats has been
present.16 In some animals, eosinophilic intracytoplasmic viral associated with formation of injection-site sarcomas.56,57 More
inclusion bodies (Negri bodies) are identified. Negri bodies are commonly, rabies virus vaccination is associated with pain at
considered a hallmark of rabies virus infection and are most the injection site, lameness, transient fever, and local cutane-
commonly seen in tissues such as the pons, thalamus, hypo- ous reactions, such as alopecia, focal cutaneous vasculitis, and
thalamus, and hippocampus. They are generally only seen once focal granulomas.58 A more generalized ischemic skin disease
neurologic signs have developed, and only in about 50% of ani- characterized by variable alopecia, crusting, erosions, and ulcers
mals that test positive using direct fluorescent antibody testing. on the pinnal margins, periocular areas, tail tip, and paw pads
Inclusions that resemble Negri bodies have been described in the has been described that occurs several months after rabies virus
brain of nonrabid cats, and so the potential for confusion and a vaccination of dogs and begins with a lesion at the site of vac-
false positive diagnosis of rabies exists.48 cination (see Chapter 12).59
138 SECTION 1  Viral Diseases

TABLE 13-4
Procedures Required by Public Health Officials for Prevention of Rabies According to the 2011 Compendium of Animal Rabies
Prevention and Control37
Situation Rabies Vaccination Procedure Rationale
History
Dog or cat exposed* Unvaccinated, dog Euthanize immediately or place in strict isolation The quarantine period is
to a rabid wild ani- or cat for 6 months. Administer rabies vaccine on based on the known incu-
mal or wild animal isolation or up to 28 days before release. bation period for rabies.
that is unavailable Not up to date Contact public health authorities, who decide pro- Animals that develop signs
for testing with booster cedures (revaccination or euthanasia) based on consistent with rabies dur-
­vaccinations biting species, local prevalence of rabies, timing ing quarantine should be
of previous vaccinations. euthanized and tested for
Vaccinated, dog or Revaccinate, and keep under owner’s control and rabies.
cat observation for 45 days.
Person bitten by a Any Confine animal and observe daily for 10 days; re- Dogs and cats shedding rabies
healthy dog or cat port illness that develops immediately to public virus within the saliva will
health authorities. If signs of rabies develop, develop illness and die
euthanize and test for rabies. Vaccination should within the 10-day quaran-
not be performed until after the isolation period tine period.
is complete.
Human bitten by a Euthanize biting animal immediately and submit
stray or unwanted for rabies testing.
dog or cat
Human bitten by a Vaccinated, human Vaccinate on day 0 and day 3 intramuscularly, in
known rabid animal the upper deltoid muscle.
Unvaccinated, Postexposure prophylaxis (injection of human
­human rabies immune globulin infiltrated at the bite
site, followed by vaccination on days 0, 3, 7, 14,
and 28)

Modified from Brown CM, Conti L, Ettestad P, et al. Compendium of animal rabies prevention and control, 2011. J Am Vet Med Assoc 2011;239:609-
617. Compendium updates are available online at http://www.nasphv.org/documentsCompendia.html.
*Exposure for rabies constitutes introduction of the virus into bite wounds, open cuts in skin, or onto mucous membranes from saliva or other po-
tentially infectious material such as neural tissue.

Oral vaccinia-vectored recombinant rabies vaccines, which the biting animal is a wild animal that is unavailable for test-
express the rabies virus glycoprotein, have been used in North ing, the biting animal is presumed to be rabid. Animals that are
America and Europe to control the spread of rabies in wild ani- overdue for a booster vaccination are evaluated on a case-by-case
mals such as raccoons, coyotes, and gray foxes.60 Canine and basis by public health authorities. Dogs and cats that are cur-
feline rabies vaccines are not approved for use in wild animals, rently vaccinated should be revaccinated immediately (within 48
which include wild animal hybrids such as wolf hybrids and hours), kept under the owner’s control, and observed for 45 days.
Savannah cats. Wolf hybrids and Savannah cats are considered Any illness in isolated or confined animals should be reported to
unvaccinated when decisions must be made regarding quaran- local public health officials. If clinical signs suggestive of rabies
tine and euthanasia (Table 13-4). develop, euthanasia and rabies testing should be performed.
Animals suspected to have rabies should be isolated from
Prevention other animals and humans and handled with extreme caution,
using heavy gloves, catch poles, and cages for restraint. Care
Prevention of rabies is dependent on widespread education of the should be taken not to allow people’s skin to come into con-
public regarding the need to vaccinate dogs and cats for rabies, tact with salivary secretions. Signs that warn of the possibility
and the risks of wildlife contact. Control of stray dogs and cats of rabies should be placed on the cages of animals that are
and wild animal vaccination programs also help to prevent the hospitalized for any period of time. If the risk of rabies is low,
disease. Ownership and importation of wild animals and hybrids the animal should be confined and observed for 10 days. The
is discouraged. Puppies and kittens that have not yet reached the clinician in charge should keep a list of all in-contact person-
age for rabies vaccination should be kept away from wildlife. nel during that period, and procedures should be minimized.
According to U.S. regulations, unvaccinated dogs and cats High-risk rabies suspects should be confined in isolation using
that have been exposed to a rabid animal should be euthanized an established hospital protocol (see Chapter 11). Marked
immediately or vaccinated and placed in strict isolation, without improvement in an animal’s condition that occurs during a
direct contact with people or other animals, for 6 months.37 If 10-day period is not consistent with a diagnosis of rabies. All
CHAPTER 13  Rabies 139

specimens submitted to the laboratory should be labeled with immediately and 3 days later. The major vaccine in use in
warning labels. the United States is the human diploid cell vaccine (HDCV),
although in developing countries, access to this vaccine is
Public Health Aspects limited. Among animal workers, antibody titers are generally
checked every 2 years, with administration of a booster dose if
Each year, 50,000 to 55,000 people die from rabies worldwide. necessary.
Approximately half those numbers occur in India alone, and it
has been estimated that 3 billion people continue to be at risk SUGGESTED READINGS
of rabies virus infection in more than 100 countries.61 A large Dietzschold B, Li J, Faber M, et  al. Concepts in the pathogenesis of
proportion of these people are children who have been attacked rabies. Future Virol. 2008;3:481-490.
by rabid dogs. In the United States, human deaths due to rabies Lackay SN, Kuang Y, Fu ZF. Rabies in small animals. Vet Clin North
have decreased from more than 100 per year in the early 20th Am Small Anim Pract. 2008;38:851-861:ix.
century to 2 to 3 per year.62 Most humans in the United States Public health response to a rabid kitten-four states, 2007. MMWR
Morb Mortal Wkly Rep. 2008;56(51-52):1337-1340.
are infected as a result of contact with insectivorous bats, which
Recovery of a patient from clinical rabies–California, 2011. MMWR
often appear injured or are behaving abnormally (including fly- Morb Mortal Wkly Rep. 2012;61:61-65.
ing during the day), and, depending on the bat species, around
5% to 40% of dead or ill bats submitted for testing have REFERENCES
rabies.63,64 Seemingly insignificant contact with tiny bats, par- 1. Adamson PB. The spread of rabies into Europe and the probable ori-
ticularly the eastern pipistrelle and silver-haired bats, is reported gin of this disease in antiquity. J R Asiat Soc GB Irel. 1977;2:140-144.
in many human rabies cases (cryptic bat rabies). Despite the 2. Lewis VJ, Thacker WL. Limitations of deteriorated tissue for rabies
high prevalence of raccoon rabies in the eastern United States, diagnosis. Health Lab Sci. 1974;11:8-12.
the only human death associated with a raccoon rabies variant 3. Matouch O, Jaros J, Pohl P. Survival of rabies virus under external
was reported in 2003.65 conditions. Vet Med (Praha). 1987;32:669-674.
Prodromal signs of rabies in humans include malaise, fever, 4. Warrell MJ, Warrell DA. Rabies and other lyssavirus diseases. Lan-
and pain, itching, or paresthesia at the site of the bite. People cet. 2004;363:959-969.
with furious rabies become nervous and hyperexcitable, after 5. Maier T, Schwarting A, Mauer D, et al. Management and outcomes
after multiple corneal and solid organ transplantations from a donor
which signs of hydrophobia, aerophobia, confusion, and
infected with rabies virus. Clin Infect Dis. 2010;50:1112-1119.
aggression develop. Autonomic signs such as hypersalivation, 6. Dietzschold B, Li J, Faber M, et al. Concepts in the pathogenesis of
vomiting, miosis or mydriasis, excessive sweating, and priapism rabies. Future Virol. 2008;3:481-490.
may occur. Paralytic rabies can be confused with Guillain-Barré 7. Davis PL, Bourhy H, Holmes EC. The evolutionary history and
syndrome.34 dynamics of bat rabies virus. Infect Genet Evol. 2006;6:464-473.
Sick domestic and wild animals that bite humans in regions 8. Fischman HR, Ward 3rd FE. Oral transmission of rabies virus in
where rabies is endemic and that subsequently die should be experimental animals. Am J Epidemiol. 1968;88:132-138.
tested for rabies without delay. Prompt and aggressive treat- 9. Jorgenson RD, Gough PM, Graham DL. Experimental rabies in a
ment of wounds including irrigation under pressure with 20% great horned owl. J Wildl Dis. 1976;12:444-447.
aqueous soap solution, and application of a greater than 50% 10. Cleaveland S, Fevre EM, Kaare M, et al. Estimating human rabies
mortality in the United Republic of Tanzania from dog bite inju-
ethanol solution or povidone-iodine, can dramatically reduce
ries. Bull World Health Organ. 2002;80:304-310.
the risk of contracting rabies after a bite. 11. Fogelman V, Fischman HR, Horman JT, et  al. Epidemiologic
The chain of events that must occur after a dog or cat bites and clinical characteristics of rabies in cats. J Am Vet Med Assoc.
a human are listed in Table 13-4 and depend on whether the 1993;202:1829-1833.
animal is owned or stray and the animal’s vaccination status.37 12. McQuiston JH, Yager PA, Smith JS, et al. Epidemiologic character-
Public health authorities must be notified when potential expo- istics of rabies virus variants in dogs and cats in the United States,
sure occurs and make the final decision as to how these animals 1999. J Am Vet Med Assoc. 2001;218:1939-1942.
are handled. 13. Blanton JD, Robertson K, Palmer D, et al. Rabies surveillance in the
Postexposure prophylaxis (PEP) is administered to humans United States during 2008. J Am Vet Med Assoc. 2009;235:676-689.
after a bite from a known or suspected rabid animal. In the 14. Castrodale L, Walker V, Baldwin J, et  al. Rabies in a puppy
imported from India to the USA, March 2007. Zoonoses Public
United States it consists of an injection of human rabies immune
Health. 2008;55:427-430.
globulin (H-RIG), ideally with at least half of the whole dose 15. Childs JE, Colby L, Krebs JW, et al. Surveillance and spatiotempo-
in the region of a bite site as soon as possible within the first 7 ral associations of rabies in rodents and lagomorphs in the United
days of the bite, followed by vaccination by the intramuscular States, 1985-1994. J Wildl Dis. 1997;33:20-27.
route on days 0, 3, 7, 14, and 28 in the upper deltoid muscle. 16. Lackay SN, Kuang Y, Fu ZF. Rabies in small animals. Vet Clin
Fewer than 5 humans develop rabies each year in the United North Am Small Anim Pract. 2008;38:851-861,ix.
States, but PEP is given to 20,000 to 40,000 people each year, 17. Eng TR, Fishbein DB. Epidemiologic factors, clinical findings, and
with on average 50 humans receiving PEP for each rabies case vaccination status of rabies in cats and dogs in the United States
investigated. in 1988. National Study Group on Rabies. J Am Vet Med Assoc.
Depending on risk, certain individuals may receive prophy- 1990;197:201-209.
18. Fekadu M, Shaddock JH, Baer GM. Excretion of rabies virus in the
lactic vaccines against rabies. These individuals include vet-
saliva of dogs. J Infect Dis. 1982;145:715-719.
erinarians, veterinary students and staff, dog catchers, rabies 19. Dimaano EM, Scholand SJ, Alera MT, et al. Clinical and epidemio-
researchers, rabies diagnosticians, wildlife workers, and travel- logical features of human rabies cases in the Philippines: a review
ers visiting rabies-enzootic regions. After exposure to a rabid from 1987 to 2006. Int J Infect Dis. 2011;15:e495-e499.
animal, administration of immunoglobulin to these individu- 20. Johnson N, Fooks A, McColl K. Human rabies case with long incu-
als is not necessary. Instead, rabies vaccine boosters are given bation, Australia. Emerg Infect Dis. 2008;14:1950-1951.
140 SECTION 1  Viral Diseases

21. Shankar SK, Mahadevan A, Sapico SD, et al. Rabies viral encepha- 42. Gupta PK, Singh RK, Sharma RN, et  al. Preliminary report on
litis with probable 25 year incubation period! Ann Indian Acad a single-tube, non-interrupted reverse transcription-polymerase
Neurol. 2012;15:221-223. chain reaction for the detection of rabies virus in brain tissue. Vet
22. Tepsumethanon V, Lumlertdacha B, Mitmoonpitak C, et al. Sur- Res Commun. 2001;25:239-247.
vival of naturally infected rabid dogs and cats. Clin Infect Dis. 43. David D, Yakobson B, Rotenberg D, et al. Rabies virus detection
2004;39:278-280. by RT-PCR in decomposed naturally infected brains. Vet Micro-
23. Murray KO, Holmes KC, Hanlon CA. Rabies in vaccinated dogs biol. 2002;87:111-118.
and cats in the United States, 1997-2001. J Am Vet Med Assoc. 44. Lopes MC, Venditti LL, Queiroz LH. Comparison between RT-
2009;235:691-695. PCR and the mouse inoculation test for detection of rabies virus
24. Fekadu M, Baer GM. Recovery from clinical rabies of 2 dogs in samples kept for long periods under different conditions. J Virol
inoculated with a rabies virus strain from Ethiopia. Am J Vet Res. Methods. 2010;164:19-23.
1980;41:1632-1634. 45. Wacharapluesadee S, Hemachudha T. Ante- and post-mortem
25. Fekadu M, Shaddock JH, Baer GM. Intermittent excretion of diagnosis of rabies using nucleic acid-amplification tests. Expert
rabies virus in the saliva of a dog two and six months after it Rev Mol Diagn. 2010;10:207-218.
had recovered from experimental rabies. Am J Trop Med Hyg. 46. Human rabies—Virginia, 2009. MMWR Morb Mortal Wkly Rep.
1981;30:1113-1115. 2010;59:1236-1238.
26. Barnes HL, Chrisman CL, Farina L, et  al. Clinical evaluation of 47. Saengseesom W, Mitmoonpitak C, Kasempimolporn S, et al. Real-
rabies virus meningoencephalomyelitis in a dog. J Am Anim Hosp time PCR analysis of dog cerebrospinal fluid and saliva samples
Assoc. 2003;39:547-550. for ante-mortem diagnosis of rabies. Southeast Asian J Trop Med
27. Gadre G, Satishchandra P, Mahadevan A, et al. Rabies viral enceph- Public Health. 2007;38:53-57.
alitis: clinical determinants in diagnosis with special reference to 48. Szlachta HL, Habel RE. Inclusions resembling Negri bodies in the
paralytic form. J Neurol Neurosurg Psychiatry. 2010;81:812-820. brains of nonrabid cats. Cornell Vet. 1953;43:207-212.
28. Laothamatas J, Wacharapluesadee S, Lumlertdacha B, et al. Furi- 49. Presumptive abortive human rabies—Texas, 2009. MMWR Morb
ous and paralytic rabies of canine origin: neuroimaging with viro- Mortal Wkly Rep. 2010;59:185-190.
logical and cytokine studies. J Neurovirol. 2008;14:119-129. 50. Recovery of a patient from clinical rabies—California, 2011.
29. Tirawatnpong S, Hemachudha T, Manutsathit S, et al. Regional dis- MMWR Morb Mortal Wkly Rep. 2012;61:61-65.
tribution of rabies viral antigen in central nervous system of human 51. Lafon M. Subversive neuroinvasive strategy of rabies virus. Arch
encephalitic and paralytic rabies. J Neurol Sci. 1989;92:91-99. Virol Suppl. 2004:149-159.
30. Centers for Disease Control and Prevention. http://www. 52. Pedersen NC, Emmons RW, Selcer R, et  al. Rabies vaccine virus
cdc.gov/rabies/specific_groups/laboratories/index.html. Last accessed infection in three dogs. J Am Vet Med Assoc. 1978;172:1092-1096.
November 19, 2012. 53. Morrison WB, Starr RM. Vaccine-associated feline sarcomas. J Am
31. Beauregard M, Boulanger P, Webster WA. The use of fluorescent Vet Med Assoc. 2001;218:697-702.
antibody staining in the diagnosis of rabies. Can J Comp Med Vet 54. Shaw SC, Kent MS, Gordon IK, et al. Temporal changes in charac-
Sci. 1965;29:141-147. teristics of injection-site sarcomas in cats: 392 cases (1990-2006).
32. Lembo T, Niezgoda M, Velasco-Villa A, et  al. Evaluation of a J Am Vet Med Assoc. 2009;234:376-380.
direct, rapid immunohistochemical test for rabies diagnosis. Emerg 55. World Health Organization. http://www.who.int/mediacentre/fact
Infect Dis. 2006;12:310-313. sheets/fs099/en/. Last accessed August 26, 2012.
33. Crepin P, Audry L, Rotivel Y, et al. Intravitam diagnosis of human 56. Kass PH, Spangler WL, Hendrick MJ, et al. Multicenter case-control
rabies by PCR using saliva and cerebrospinal fluid. J Clin Micro- study of risk factors associated with development of vaccine-associated
biol. 1998;36:1117-1121. sarcomas in cats. J Am Vet Med Assoc. 2003;223:1283-1292.
34. Madhusudana SN, Sukumaran SM. Antemortem diagnosis and pre- 57. Srivastav A, Kass PH, McGill LD, et  al. Comparative vaccine-­
vention of human rabies. Ann Indian Acad Neurol. 2008;11:3-12. specific and other injectable-specific risks of injection-site sarcomas in
35. Li Z, Feng Z, Ye H. Rabies viral antigen in human tongues and cats. J Am Vet Med Assoc. 2012;241:595-602.
salivary glands. J Trop Med Hyg. 1995;98:330-332. 58. Wilcock BP, Yager JA. Focal cutaneous vasculitis and alope-
36. Jogai S, Radotra BD, Banerjee AK. Rabies viral antigen in extracra- cia at sites of rabies vaccination in dogs. J Am Vet Med Assoc.
nial organs: a post-mortem study. Neuropathol Appl Neurobiol. 1986;188:1174-1177.
2002;28:334-338. 59. Vitale CB, Gross TL, Magro CM. Vaccine-induced ischemic der-
37. Brown CM, Conti L, Ettestad P, et  al. Compendium of ani- matopathy in the dog. Vet Dermatol. 1999;10:131-142.
mal rabies prevention and control, 2011. J Am Vet Med Assoc. 60. Sidwa TJ, Wilson PJ, Moore GM, et al. Evaluation of oral rabies
2011;239:609-617. vaccination programs for control of rabies epizootics in coyotes and
38. Moore SM, Hanlon CA. Rabies-specific antibodies: measuring sur- gray foxes: 1995-2003. J Am Vet Med Assoc. 2005;227:785-792.
rogates of protection against a fatal disease. PLoS Negl Trop Dis. 61. Wunner WH, Briggs DJ. Rabies in the 21 century. PLoS Negl Trop
2010;4:e595. Dis. 2010;4:e591.
39. Panning M, Baumgarte S, Pfefferle S, et al. Comparative analysis 62. Nigg AJ, Walker PL. Overview, prevention, and treatment of
of rabies virus reverse transcription-PCR and virus isolation using rabies. Pharmacotherapy. 2009;29:1182-1195.
samples from a patient infected with rabies virus. J Clin Microbiol. 63. Balsamo GA, Ratard RC. Epidemiology of animal rabies and its
2010;48:2960-2962. practical application to pre- and postexposure prophylaxis, Louisiana,
40. Fooks AR, Johnson N, Freuling CM, et al. Emerging technologies 1988 to 2007. Vector Borne Zoonotic Dis. 2010;10:283-289.
for the detection of rabies virus: challenges and hopes in the 21st 64. Shankar V, Orciari LA, De Mattos C, et al. Genetic divergence of
century. PLoS Negl Trop Dis. 2009;3:e530. rabies viruses from bat species of Colorado. USA. Vector Borne
41. Kamolvarin N, Tirawatnpong T, Rattanasiwamoke R, et al. Diag- Zoonotic Dis. 2005;5:330-341.
nosis of rabies by polymerase chain reaction with nested primers. 65. First human death associated with raccoon rabies—Virginia, 2003.
J Infect Dis. 1993;167:207-210. MMWR Morb Mortal Wkly Rep. 2003;52:1102-1103.
CHAPTER 14
Canine Parvovirus Infections and
Other Viral Enteritides
Jane E. Sykes

is thought to have minimal pathogenic potential, but it has been


Overview of Canine Parvoviral Enteritis associated with abortion in pregnant dogs; respiratory, cardiac,
First Described: 1978, worldwide, Appel and others1 and gastrointestinal signs in neonatal dogs; severe gastroenteri-
Cause: Canine parvovirus-2 variants (Family Parvoviridae, tis in adult dogs; and possibly neurologic signs in dogs.6-8
subfamily Parvovirinae, Genus Parvovirus) Parvoviruses are small, non-enveloped single-stranded DNA
viruses (Figure 14-1, A) that can survive for long periods (over
Affected Hosts: Dogs and other Canidae such as coyotes,
1 year) in the environment. As a result, contact with virus that
foxes, and wolves; cats
persists in the environment is an important means of transmis-
Geographic Distribution: Worldwide sion. Insects and rodents may also serve as mechanical vectors
Route of Transmission: Direct contact with virus in feces and for the virus. Canine parvovirus requires the presence of mitoti-
vomitus as well as contact with contaminated fomites cally active cells in order to replicate. Young animals (6 weeks
to 6 months, and especially those less than 12 weeks of age) are
Major Clinical Signs: Fever, lethargy, inappetence, vomiting,
more likely to develop severe illness; however, disease can also
diarrhea, dehydration. Sudden death or tachypnea due to
occur in unvaccinated or improperly vaccinated adult dogs. In
myocarditis occurs rarely.
North America, Rottweilers, American pit bull terriers, Dober-
Differential Diagnoses: Canine distemper virus infection, man pinschers, English springer spaniels, and German shep-
other canine viral enteritides, dietary indiscretion, toxins, herd dogs appear to be at increased risk for development of
gastrointestinal foreign body, enteric parasitic infections parvoviral enteritis,9,10 but this has not been the case in other
such as giardiasis and helminth infections, enteric bacte- geographic locations. A seasonal distribution of disease has
rial infections such as salmonellosis, salmon poisoning been reported in some geographic locations, which may reflect
disease, pancreatitis, hypoadrenocorticism, inflammatory times when dogs access the outdoors and contact virus in the
bowel disease environment. For example, in Saskatoon, Canada, dogs were
Human Health Significance: CPV-2 variants do not infect three times more likely to be admitted with parvoviral enteritis
humans in July, August, and September, compared with the rest of the
year.9 In other locations, the seasonal pattern of disease has dif-
fered or been nonexistent.11,12 For dogs older than 6 months
of age, intact males were twice as likely as females to develop
Etiology and Epidemiology parvoviral enteritis.9 A surveillance study from Australia found
a correlation between clusters of parvoviral enteritis and regions
Canine parvovirus is the most widely recognized cause of trans- of relative socioeconomic disadvantage.12
missible viral diarrhea in dogs and one of the most common Other viral pathogens that have been associated with enteri-
infectious diseases of dogs worldwide. It is caused by variants of tis and diarrhea in dogs are canine distemper virus (CDV)
canine parvovirus-2 (CPV-2), which are members of the genus (see Chapter 15), canine enteric coronavirus (Figure 14-1, B),
Parvovirus. CPV-2 emerged in the early to mid-1970s and rotaviruses, astroviruses, adenoviruses, caliciviruses, and
caused a worldwide pandemic of illness in dogs.2 The spread of novel viruses that include a norovirus, kobuvirus, sapovirus,
the virus worldwide occurred over a remarkable period of about and possibly also a circovirus.13-17 Canine enteric coronavirus
6 months. CPV-2 may have been derived from feline panleuko- primarily causes mild diarrhea in puppies that are less than
penia virus (FPV) or a closely related virus of wild carnivores. 6 weeks of age and may be found in co-infections with other
It has since mutated to CPV-2a in 1979, CPV-2b in 1984, and, viral causes of gastroenteritis, including CPV-2 variants. Rarely,
most recently, CPV-2c, which was first detected in Italy in 2000 it has been identified as a more significant cause of diarrhea in
and has subsequently been found worldwide, with the exception young dogs.18 This chapter focuses on canine parvoviral enteri-
of Australia. Separate lineages have been identified in different tis, which is the most widely recognized and pathogenic viral
geographic locations worldwide.3 The virus uses the transferrin enteritis of dogs.
receptor to enter host cells. With the mutation from CPV-2 to
CPV-2a, the virus developed the ability to replicate readily in Clinical Features
feline cells, and CPV-2a, CPV-2b, and CPV-2c are now respon-
sible for some cases of feline viral enteritis.4,5 Feline parvoviral Signs and Their Pathogenesis
enteritis is further discussed in Chapter 19. CPV-2 variants must Transmission of parvovirus and other viral causes of gastroen-
be differentiated from CPV-1, also known as canine minute teritis occurs by the fecal-oral route, after exposure to virus in
virus, which belongs to the genus Bocavirus. In general, CPV-1 feces or vomit, or importantly, virus that persists on fomites.

141
142 SECTION 1  Viral Diseases

Spike glycoprotein

Helical
nucleocapsid

A B
FIGURE 14-1  A, General structure of canine parvovirus. The virus is non-enveloped,
is 25 nm in diameter, and has icosahedral symmetry. B, Structure of a coronavirus, which
is an enveloped virus. Canine parvovirus is about one-quarter the size of a coronavirus.

Virus is shed for a few days before the onset of clinical signs,
and shedding declines considerably after 7 days.19 The severity
of clinical signs depends on factors such as virus strain and host
immunity, which is affected by stressors such as weaning and
overcrowding, maternal antibody, and the presence of concur-
FIGURE 14-2  Target sites of replication of selected enteric viral pathogens. The most
rent infections such as other enteric viral and parasitic infec-
pathogenic enteric viruses, such as canine parvovirus, replicate and destroy crypt epithelial
tions. Subclinical infections are probably widespread. cells.
The incubation period for canine parvoviral enteritis is 7 to
14 days in the field, but shorter incubation periods (as short as 4
days) have been observed with experimental infections. The virus this period, the incidence of neonatal complications of parvo-
replicates in oropharyngeal lymphoid tissues, after which viremia virus infection and myocarditis has declined dramatically since
occurs. Damage occurs to rapidly dividing cells in the gastroin- the virus first emerged, because of widespread vaccination and
testinal tract, thymus, lymph nodes, and bone marrow. Affected exposure of adult animals.
gastrointestinal tissues include the epithelium of the tongue, oral Neurologic signs in puppies with parvoviral enteritis may
cavity, esophagus, and intestinal tract, and especially the germi- result from hypoxia secondary to myocarditis, hypoglycemia,
nal epithelial cells of the intestinal crypts. Neutropenia results or intracranial thrombosis or hemorrhage. The possibility of
not only from infection of the marrow but also sequestration of co-infection with CDV should also be considered. The DNA
neutrophils in damaged gastrointestinal tissue. Malabsorption of CPV-2 variants has also been detected in the central nervous
and increased intestinal permeability result. Secondary bacterial system,26 and there have been rare reports of leukoencephalo-
infections of the gastrointestinal tract, which may be followed malacia in association with infection.27,28
by bacterial translocation, bacteremia, and endotoxemia, play In contrast to canine CPV-2 variants, the replication of less
a key role in the pathogenesis of the disease. Mucosal candidia- pathogenic enteric viruses is restricted to the intestinal tract,
sis has also been described in puppies with parvoviral enteritis and the crypt epithelial cells are generally spared (Figure 14-2).
(see Chapter 67). The outcome is clinical signs of fever, leth- Canine enteric coronavirus, for example, infects the mature
argy, inappetence, vomiting, diarrhea, rapid dehydration, and enterocytes at the tips of the villi. Crypt cell hyperplasia occurs
abdominal pain. Diarrhea is often liquid, foul-smelling, and may to replace the damaged cells. The villi become shortened or dis-
contain streaks of blood or frank blood. Ascarid nematodes may torted, which leads to malabsorption and diarrhea.
be identified in the vomitus of some dogs. Dogs with canine par-
voviral enteritis have evidence of disordered coagulation, with Physical Examination Findings
decreased antithrombin activities, prolonged activated partial Physical examination of puppies with parvoviral enteritis often
thromboplastin time, increased thromboelastography maximum reveals fever (up to 41°C or 105°F), lethargy, weakness, dehy-
amplitude, and increased fibrinogen concentrations. Catheter dration, and abdominal tenderness and a fluid-filled intestinal
and organ thrombosis may occur.20 Secondary bacteremia may tract on palpation (Figure 14-3). Vomiting or diarrhea may
be associated with multiple organ failure and death. occur during the examination, or there may be evidence of diar-
Infection of the dam by CPV-2 variants early in gestation rhea or frank blood on the perineum or the rectal thermometer.
can lead to infertility, resorption, or abortion. Puppies that are Occasionally abdominal palpation reveals a tubular mass as a
infected in utero or up to 2 weeks of age may develop viral result of intestinal intussusception. Ulcerative glossitis can occur
myocarditis, which results in signs of sudden death or conges- in some puppies. Mucosal pallor, prolonged capillary refill time,
tive heart failure.21-23 Damage to the developing myocardium or rarely hypothermia may be observed in some dogs.29 Septic
usually occurs up to the first 2 weeks of life, but clinical signs of shock may be associated with tachycardia or bradycardia, men-
myocardial damage may be delayed until up to 2 months of age. tal obtundation, and poor pulse quality. Uncommonly, neuro-
Cerebellar hypoplasia has been rarely reported in dogs after in logic signs such as tremors and seizures are observed. Puppies
utero infection24 but is more common in kittens infected with with myocarditis may be tachypneic and have increased lung
FPV (see Chapter 19). Generalized infection has been reported sounds as a result of congestive heart failure. Erythema multi-
in neonatal puppies, with hemorrhage and necrosis within the forme has been reported in dogs with canine parvoviral enteritis;
brain, liver, lungs, kidneys, lymphoid tissues, and gastrointesti- these dogs had generalized cutaneous and mucosal ulceration as
nal tract.25 Because maternal antibody protects puppies during well as swelling of the pinnae and paws.30,31
CHAPTER 14  Canine Parvovirus Infections and Other Viral Enteritides 143

Diagnosis
Laboratory Abnormalities
Complete Blood Count
The most common abnormalities found on the CBC are leuko-
penia, neutropenia, and lymphopenia (Table 14-1). Toxic neu-
trophils and monocytopenia may also be present. Only around
one third of dogs had leukopenia in one study.29 Leukopenia
can develop after the onset of gastrointestinal signs, when
puppies are first brought for examination. Some dogs have
leukocytosis due to a neutrophilia and monocytosis. Although
the presence of leukopenia supports a diagnosis of parvoviral
enteritis, other severe gastrointestinal infections such as sal-
monellosis can also cause leukopenia and diarrhea, so it is not
specific for diagnosis of the disease. Thrombocytosis or, less
commonly, thrombocytopenia may also occur.29 Some pup-
pies develop anemia as a result of gastrointestinal blood loss,
which may be non regenerative or become regenerative.

Serum Biochemical Tests


The serum biochemistry panel in dogs with parvoviral enteritis
often shows hypoproteinemia, hypoalbuminemia, and hypogly-
cemia. Mild hyperglycemia has also been reported. Electrolyte
abnormalities such as hyponatremia, hypochloremia, and hypo-
kalemia may occur (Table 14-2). Occasionally severe dehydration
results in prerenal azotemia. Puppies with bacterial sepsis may
develop increased liver enzyme activities and hyperbilirubinemia.

Coagulation Profile
Coagulation abnormalities have been reported in a small num-
FIGURE 14-3  Obtundation in a 3-month-old intact male standard poodle puppy ber of dogs with parvoviral enteritis. When abnormalities occur,
with canine parvovirus enteritis. The dog had been vaccinated for canine parvovirus at 8 findings include prolonged activated partial thromboplastin
and 10 weeks of age. time, decreased antithrombin activity, increased fibrinogen

TABLE 14-1
Complete Blood Count Findings at Admission in 45 Dogs Diagnosed with Canine Parvoviral Enteritis at the UC Davis VMTH
Percent below Percent within Percent above Range for
Reference the Reference the Reference the Reference Dogs with CPV Number
Test Range Range Range Range Enteritis Tested
Hematocrit (%) 40-55 71 29 0 21-53 45
MCV (fL) 65-75 29 71 0 54-74 45
MCHC (g/dL) 33-36 24 69 7 30-37 45
Neutrophils (cells/µL) 3000-10,500 56 31 13 8-22,453 45
Band neutrophils 0-rare 0 31 69 0-1582 45
(cells/µL)
Monocytes (cells/µL) 150-1200 20 60 20 11-2475 45
Lymphocytes (cells/µL) 1000-4000 49 22 0 165-3698 45
Eosinophils (cells/µL) 0-1500 0 100 0 0-1236 45
Platelets (cells/µL) 150,000-400,000 5 60 35 103,000- 43
639,000

Note: Adult reference ranges were used by the laboratory.


CPV, Canine parvovirus.
144 SECTION 1  Viral Diseases

TABLE 14-2
Findings on Serum Biochemistry Analysis in 27 Dogs with Canine Parvoviral Enteritis at the UC Davis VMTH
Percent below Percent within Percent above Range for Dogs
Reference the Reference the Reference the Reference with Parvoviral Number of
Test Range Range Range Range Enteritis Dogs Tested
Sodium (mmol/L) 145-154 85 15 0 132-147 26
Potassium (mmol/L) 3.6-5.3 4 92 4 3.4-5.9 26
Chloride (mmol/L) 108-118 65 31 4 94-120 26
Bicarbonate (mmol/L) 16-26 8 92 0 14-25 26
Calcium (mg/dL) 9.7-11.5 30 70 0 7.4-11.5 27
Phosphorus (mg/dL) 3.0-6.2 0 52 48 3.1-12.3 27
Creatinine (mg/dL) 0.3-1.2 37 59 4 <0.2-1.6 27
BUN (mg/dL) 5-21 0 85 15 5-48 27
Albumin (g/dL) 3.0-4.4 70 30 0 0.9-3.8 27
Globulin (g/dL) 1.8-3.9 26 67 7 1.4-4.5 27
Cholesterol (mg/dL) 135-361 4 89 8 108-460 26
Total bilirubin (mg/dL) 0-0.2 0 89 12 0-1.7 26
ALT (U/L) 19-67 15 62 23 10-165 26
ALP (U/L) 21-170 0 42 58 56-397 26

Note: Adult reference ranges were used by the laboratory.

concentrations, and increased thromboelastography maximum although they detect all variants of CPV-2 including CPV-2c,
amplitude.20 An increase in D-dimers or fibrin degradation their sensitivities and specificities vary.32,33 Sensitivity is par-
products has not been reported. More research is warranted ticularly problematic, because viral shedding is transient, and
to understand the range of coagulation abnormalities that can antibody present may bind viral antigen so that it is unavail-
occur in parvoviral enteritis. able for reaction with the assay. The sensitivities of three com-
mercially available fecal antigen assays (SNAP Parvo antigen,
Diagnostic Imaging IDEXX Laboratories GmbH; FASTest Parvo Strip, Scil Ani-
Plain Radiography mal Care Company GmbH; Witness Parvo Card, Selectavet
Plain abdominal radiography in dogs with parvoviral enteritis GmbH) were 50%, 40%, and 60%, respectively, when com-
may show poor serosal detail (often due to lack of intra-abdom- pared to immunoelectron microscopy, and 18%, 16%, and
inal fat in puppies) and a fluid- and gas-filled gastrointestinal 26%, respectively, when compared with results of a fecal PCR
tract. Abdominal radiography is usually performed to assess for assay.33 Because some dogs that lack evidence of gastrointes-
the presence of a gastrointestinal foreign body. tinal signs can be positive using PCR assay, immunoelectron
microscopy may be a more appropriate gold standard for dis-
Sonographic Findings ease (although it is not widely available for routine diagno-
Findings on abdominal ultrasonography in canine parvoviral sis). In another study, the sensitivity of a fecal antigen test for
enteritis are nonspecific but can include a thickened gastroin- detection of CPV-2a, CPV-2b, and CPV-2c, on specimens that
testinal mucosa, mild peritoneal effusion, fluid distention of the contained high CPV DNA loads (>105 copies/mg of feces as
gastrointestinal tract, and decreased gastrointestinal motility. determined with real-time PCR) was 80%, 78%, and 77%,
Mild mesenteric lymphadenopathy may be present. Abdominal respectively.32
ultrasound is useful to confirm a diagnosis of secondary intesti- When compared with PCR and immunoelectron microscopy
nal intussusception. of stool, the specificities of the three commercially available
antigen assays above were 98%, 98%, and 92%, respectively,33
Microbiologic Tests so false positives were uncommon. It has been suggested that
Diagnostic assays for canine parvoviral enteritis in dogs are false-positive antigen tests can occur 4 to 8 days after vacci-
listed in Table 14-3. nation with attenuated live CPV-2 vaccines.34 In kittens, false-
positive test results did occur after vaccination for FPV, but
Fecal Parvovirus Antigen ELISA they were more likely to occur with some assays as opposed to
The most widely used assay for diagnosis of canine parvoviral others, were generally weak positives, and occurred even after
enteritis is an in-house fecal antigen ELISA, which is performed vaccination with inactivated vaccines.35 Similar studies have not
on a rectal swab specimen. Several assays are available, and been reported for CPV.
CHAPTER 14  Canine Parvovirus Infections and Other Viral Enteritides 145

TABLE 14-3
Diagnostic Assays Available for Canine Parvoviral Enteritis in Dogs
Assay Specimen type Target Performance
Fecal antigen Feces CPV antigen Specificity for detection of virus nears 100%, but weak
ELISA false positives have the potential to occur after immu-
nization with attenuated live vaccines. False negatives
are common (low sensitivity).
Hemagglutina- Feces CPV antigen Inexpensive and rapid. Sensitivity and specificity in natu-
tion assay rally infected dogs has not been well established.
Histopathology Usually necropsy Crypt necrosis with Can be used for diagnosis at necropsy. In situ hybridiza-
specimens, especial- intranuclear inclusions; tion may be most sensitive for detection of virus in
ly gastrointestinal parvovirus antigen tissues.
tissues with IHC or parvovi-
rus DNA with in situ
hybridization
Polymerase chain Feces, tissue species CPV DNA Sensitivity and specificity may vary depending on assay
reaction (PCR) design. Attenuated live vaccine virus may be detected
in feces for days to weeks after vaccination, but some
assays differentiate between vaccine and field virus.
Because of the high sensitivity of some assays, the
significance of a positive result may be difficult to
interpret. False negative results may occur as a result
of inhibition of PCR by components of feces. Degrada-
tion of nucleic acid during specimen transport is more
problematic for RNA viruses such as canine coronavi-
rus.
Fecal electron Feces Virus particles Not widely available, turnaround time can be slow,
microscopy and may be expensive. Requires the presence of large
amounts of virus.
Virus isolation Feces, tissues CPV Difficult, not widely available. Used as a research tool.

CPV, Canine parvovirus (refers to CPV-2 variants); IHC, immunohistochemistry.

Hemagglutination Testing dogs with chronic diarrhea, and so it may be difficult to ascer-
Canine parvovirus agglutinates erythrocytes, and so the presence tain whether a positive parvovirus PCR result indicates that
of the virus in stool can be detected with a simple hemagglutina- CPV is the cause of a dog’s illness. Attenuated live vaccine virus
tion test that involves mixing a suspension of feces with porcine can also be detected in the feces with PCR assays after vaccina-
erythrocytes. Agglutination of erythrocytes in a microwell plate tion, although assays have been designed that can differentiate
or on a slide indicates the presence of parvovirus in the feces.36 between vaccine and wild-type virus.37 It is not yet clear for how
The sensitivity and specificity of these assays in dogs with and long after vaccination false-positive PCR results might occur,
without natural parvoviral infections in the field require further and this could vary based on the design of the PCR assay used.
investigation. Both vaccine and field virus have been detected simultaneously
in some dogs using these assays. In the future, quantitation of
Molecular Diagnosis Using the Polymerase Chain Reaction virus loads in feces using real-time PCR may be helpful for inter-
Several commercial veterinary diagnostic laboratories now offer pretation of the significance of a positive PCR assay result.
real-time PCR assays for detection of CPV-2 variants and other
enteric viral pathogens (such as canine coronavirus). Assays Fecal Electron Microscopy
may detect as few as 1000 copies of viral DNA per milligram of Fecal electron microscopy is still offered by some institutions
stool. Turnaround times are less than 24 hours in some labora- for diagnosis of viral enteritis. It is generally used on a research
tories. PCR assays are useful when fecal antigen tests are nega- basis when viral enteritis is suspected but a diagnosis cannot be
tive but parvoviral enteritis is still suspected as a diagnosis, or made with antigen tests or PCR assays. Fecal electron micros-
when canine enteric coronavirus infection is a potential cause of copy may facilitate diagnosis of other viral infections such as
illness (because PCR panels that assay for parvovirus DNA also rotavirus, norovirus, and coronavirus infections. Turnaround
often include an assay for canine enteric coronavirus RNA). time may be slow. Generally speaking, large amounts of virus
Unfortunately, although infrequent, positive PCR assay results must be present for results to be positive, and technical expertise
for CPV can occur in dogs without signs of gastroenteritis or in is required to accurately identify virus in the stool.
146 SECTION 1  Viral Diseases

can also be present. Rarely, central nervous system lesions con-


sisting of leukoencephalomalacia have been described.27 Viral
intranuclear inclusions may be visible in some cells, especially
the intestinal crypt epithelium. Immunohistochemistry can be
used to detect viral antigen in the gastrointestinal tract, mar-
row, lymphoid tissues, and rarely in the myocardium. In situ
hybridization (see Chapter 5) can also be used to detect virus in
histopathology specimens and may have greater sensitivity than
immunohistochemistry.39,40

Treatment and Prognosis


Antimicrobial Treatment and Supportive Care
Treatment of CPV enteritis involves supportive care and treat-
ment of secondary bacterial infections with antimicrobial drugs
(Table 14-4). Whenever possible, the patient should be hospi-
talized in isolation. Appropriate fluid therapy and maintenance
of adequate blood glucose concentrations are the most critical
FIGURE 14-4  Discoloration of the small intestinal wall and serosal hemorrhage in aspect of treatment. Whenever possible, fluids should be given
a puppy that died of canine parvoviral enteritis. (Courtesy University of California, Davis intravenously and supplemented as needed with potassium
Veterinary Anatomic Pathology Service.) chloride and dextrose. Blood glucose concentrations should
be monitored at least twice daily, and more frequent monitor-
ing may be indicated if hypoglycemia is present. Although not
Virus Isolation generally recommended for puppies that are vomiting or dehy-
Canine parvovirus variants can be isolated in canine and feline drated, administration of subcutaneous fluids and antimicrobial
cells, but isolation is difficult, and the virus shows minimal drugs in the home can sometimes result in recovery when client
cytopathic effects. As a result, virus isolation is rarely used for finances do not permit treatment in hospital. Fluids adminis-
diagnosis and is not widely available. It remains important as a tered subcutaneously should never be supplemented with dex-
research tool.38 trose, because dextrose is hyperosmotic and can cause further
dehydration, as well as injection-site reactions. Unfortunately,
Serology the owners of many dogs with parvoviral enteritis lack finan-
Antibodies to CPV-2 can be measured in the laboratory using cial resources for treatment. In these situations, the inability to
hemagglutination inhibition (see Chapter 2). In addition, an afford vaccination may have been a reason the puppy develops
in-clinic ELISA assay is available for semiquantitative measure- parvoviral enteritis in the first place.
ment of antibodies to CPV-2. These assays are generally used An antimicrobial drug or drug combination with activity
to assess the need for vaccination, rather than for diagnosis of against gram-negative and anaerobic bacteria should be admin-
CPV-2 enteritis, because affected dogs are either seronegative istered parenterally. Injectable ampicillin or cefazolin alone may
or previous vaccination or material antibodies confound early be sufficient for many dogs, but puppies that have hemorrhagic
serodiagnosis. diarrhea or evidence of the systemic inflammatory response syn-
drome (SIRS) should probably be treated with a combination of
Pathologic Findings a penicillin and a fluoroquinolone, or a combination of a peni-
Gross Pathologic Findings cillin and an aminoglycoside. Use of fluoroquinolones in young,
Gross pathologic findings in dogs with CPV enteritis include rapidly growing animals has been associated with cartilage
thickening and discoloration of the intestinal wall with serosal damage (see Chapter 8), but when used for the short periods of
hemorrhage (Figure 14-4) and enlarged, edematous abdominal time required to treat parvoviral enteritis, this may not be of sig-
lymph nodes. The intestine may contain bloody liquid con- nificant concern. Proper hydration is critical before aminogly-
tents, and mucosal hemorrhage may be identified. Pale areas cosides are used because of their potential for nephrotoxicity.
may be seen within the myocardium of dogs with parvoviral Treatment with antiemetics (such as a constant rate infusion
myocarditis. of metoclopramide or parenteral ondansetron), H2 blockers
such as famotidine, whole blood or plasma transfusions, col-
Histopathologic Findings loids such as hetastarch, or partial or total parenteral nutrition
The major histopathologic finding is necrosis of the crypt epi- may be indicated in some dogs. Placement of a central line or
thelium in the small intestine, with widespread systemic lym- multilumen catheter may be necessary in severely ill puppies,
phoid depletion and necrosis. The crypts can be dilated and but strict sterile technique must be adhered to because of the
distended with cellular debris and mucus (Figure 14-5). Pro- potential for hospital-associated infection. In general, unless
liferation of crypt enterocytes may be observed as part of the absolutely necessary, invasive surgical procedures and the use
recovery response. Intestinal villi are collapsed, shortened, and of parenteral nutrition solutions should be avoided in puppies
fused, with attenuation of the epithelial lining, and there may be with severe neutropenia. Whether plasma or hetastarch is the
mild to severe fibrinous inflammation and hemorrhage. Myeloid treatment of choice for dogs with low colloid oncotic pres-
depletion may be found in the bone marrow. Parvoviral myocar- sure requires further study. Plasma may offer benefit over het-
ditis is characterized by myocardial degeneration and necrosis, astarch in that it contains antibodies from immune dogs, but
with a lymphocytic inflammatory infiltrate. Myocardial fibrosis titers may not be sufficient to be beneficial, and most puppies
CHAPTER 14  Canine Parvovirus Infections and Other Viral Enteritides 147

A B
FIGURE 14-5  A, Segmental crypt necrosis in the ileum of a dog infected with a CPV-2 variant. There is severe loss of crypt epithelial cells. Hematoxylin and eosin (H&E) stain.
B, Jejunum of a dog after infection by a CPV-2 variant. Regenerating epithelial cells, which here are nested in an inflamed jejunal lamina, have a large and bizarre appearance and resemble
adenoma cells. As a result, the disease has been termed adenomatosis. Parvovirus antigen was no longer detectable by immunohistochemistry at this stage. The crypts in the bottom left
corner have a more normal appearance. H&E stain. (Courtesy Dr. Patricia Pesavento, University of California, Davis Veterinary Anatomic Pathology Service.)

been associated with acute kidney injury in critically ill human


TABLE 14-4 patients.41 Early enteral nutrition with a nasogastric feeding
Medications That May Be Used in Conjunction with Fluid tube was associated with reduced hospitalization times in one
study, as compared to withholding food until vomiting had
Therapy to Treat Canine Parvoviral Enteritis
ceased (Figure 14-6).42 Gastric suction should be performed
through the tube before food is administered.
Interval
Other treatments that have been investigated include anti-
Drug Dose (mg/kg) Route (hours)
endotoxin sera, recombinant bactericidal permeability-increasing
Ampicillin sodium 20 IV 6 (BPI) protein, recombinant granulocyte colony-stimulating
Cefazolin sodium 20 IV 8 factor (G-CSF), recombinant feline interferon-omega (rfIFN-ω,
Enrofloxacin* 5 IV 24 Virbagen omega), and oseltamivir. Anti-endotoxin sera, recom-
binant BPI protein, and human recombinant G-CSF were
Ondansetron 0.5 to 1 IV 12 not found to be beneficial. Although neutrophil counts were
Maropitant citrate 1 SC 24 higher and hospital times were shorter in dogs treated with
Metoclopramide 1-2 mg/kg/d IV CRI recombinant canine G-CSF, survival times in these dogs were
decreased.43 Treatment of parvovirus infections with G-CSF
Famotidine 0.5 IV 12 to 24
might cause harm, because the increased cell turnover induced
CRI, Constant-rate infusion. by the drug might promote parvovirus replication. Treatment of
*Has been associated with cartilage injury in growing animals. Pro- puppies with parvoviral enteritis with rfIFN-ω in a number of
longed use (>7 days) is not recommended. See Chapter 8. placebo-controlled trials has been associated with reduced dis-
ease severity and, in some studies, significantly reduced mortal-
ity, but it has not been beneficial for treatment of FPV infections
show evidence of an antibody response within 3 days after onset (see Chapter 19).
of clinical signs. Hetastarch has anticoagulant properties that Oseltamivir inhibits the neuraminidase of influenza viruses
could be beneficial in light of the hypercoagulable state that has and does not have specific anti-parvoviral activity, but it has
been documented in canine parvoviral enteritis, but these effects been widely used for treatment of canine parvoviral enteritis. As
also have the potential to increase mortality, and hetastarch has canine parvovirus does not possess a neuraminidase, it has been
148 SECTION 1  Viral Diseases

Prognosis
The prognosis for viral enteritis in puppies varies with the severity
of illness and the owners’ ability to afford appropriate treatment.
Survival rates for puppies with CPV-2 enteritis have ranged from
9% in untreated puppies to greater than 90% with aggressive
treatment in tertiary referral hospitals.10,45-47 Factors that have
been related to mortality include the presence of initial leukope-
nia or lymphopenia, monocytopenia, neutropenia, and evidence
of SIRS.29,48,49 SIRS was defined as the presence of at least three of
the following four criteria: heart rate greater than 140 beats/min,
temperature above 102.5°F or below 100°F, and white blood
cell counts greater than 17,000 or less than 6000 cells/µL (see
Chapter 86). Survival may also be lower in Rottweilers when
compared to other breeds and in young puppies (less than 7 to
12 weeks of age).50 In Australia, euthanasia was more likely to
occur in summer; among pedigree dogs, in hounds, gundogs, and
non-sporting breed dogs; and in puppies less than 6 months of
age.47 Vomiting and lethargy at admission as well as lympho-
penia and hypoalbuminemia have been associated with pro-
longation of hospitalization times by approximately 2 days.29
Positive changes in leukocyte counts (and especially lymphocyte
counts) as early as 24 hours after admission have also been asso-
ciated with survival.48 In general, puppies that survive the first
3 to 4 days of treatment make complete recoveries. Complications
of infection include bacteremia and septic shock, intestinal intus-
susception, aspiration pneumonia, and esophageal strictures.

Immunity and Vaccination


Immunity that follows natural infection by CPV-2 variants is
FIGURE 14-6  Puppy in Figure 14-2 after placement of a nasogastric feeding tube. probably lifelong. Immunization with attenuated live viral vac-
cines also provides sterile immunity that may even be lifelong.
Both attenuated live and inactivated CPV vaccines are available.
With the possible exception of shelter environments, attenuated
hypothesized that oseltamivir instead may act on the neuramini- live vaccines should never be administered to pregnant bitches
dases of bacteria that are normally responsible for secondary because they may cause disease in the developing fetus. If possible,
bacterial infections in parvovirus enteritis, primarily those of the pregnant bitches introduced into shelter environments should be
gastrointestinal tract. A single prospective, randomized, masked, tested for antibody to CPV before vaccination with attenuated live
placebo-controlled trial of 35 dogs with parvovirus enteritis vaccines, and these dogs should only be vaccinated if they test
showed that dogs treated with oseltamivir (2 mg/kg PO q12h) negative. The use of inactivated vaccines is not recommended in
had no significant drop in their leukocyte count, whereas contaminated environments, because the window of vulnerability
untreated dogs had a significant drop in their leukocyte count and the time to onset of protection is too long. The window of
in the first 5 days of hospitalization.44 Treated dogs also vulnerability is the period when maternal antibody interferes with
gained weight during hospitalization, whereas untreated dogs the vaccine’s ability to stimulate an effective immune response,
lost weight. However, there was no difference in hospitaliza- but does not prevent infection with virulent field virus (see Chap-
tion time, clinical scores, morbidity, or mortality between the ter 12 for a discussion of this concept). Small quantities of attenu-
two groups, and the number of dogs in each group was small. ated live vaccine virus are shed from the intestinal tract after
The authors acknowledged the potential concerns that relate to immunization, but this should not be relied on to immunize other
administration of an oral medication to dogs with enteritis, with in-contact animals. Protection against challenge with field virus
possible variability in drug absorption. A major concern that occurs for at least 3 years and possibly for life after the initial
relates to treatment of canine parvoviral enteritis with oselta- puppy series is administered (see Chapter 12). Exposure of dogs
mivir is the possibility of selection for resistant mutants among to virus in the environment may also serve to booster immunity
influenza viruses if widespread use of the drug occurs in veteri- after vaccination.
nary clinics. Given the restrictions on the use of this drug for The initial immunization series should be given every 3 to 4
treatment of human influenza virus infections, further investiga- weeks from 6 to 8 weeks of age, until no sooner than 14 to 16
tion is required before the widespread use of oseltamivir can be weeks (16 to 20 weeks in breeding kennels), because of persis-
recommended for treatment of CPV enteritis. More informa- tence of sufficient titers of maternal antibody in some puppies
tion on canine G-CSF, rfIFN-ω, and oseltamivir can be found in until this age. After that, a booster should be given at 1 year
Chapter 7 of this book. of age, and every 3 years thereafter (see Appendix ). Puppies
After recovery from viral enteritis, intestinal parasites should be isolated in the home environment until 7 to 10 days
should be treated with a broad-spectrum anthelmintic such as after the last booster. Attendance at organized puppy classes has
fenbendazole. been advocated as a way to promote socialization in this period,
CHAPTER 14  Canine Parvovirus Infections and Other Viral Enteritides 149

and currently there is no evidence that such activities increase glutaraldehyde and ortho-phthalaldehyde, when contact times
risk for parvoviral enteritis. Serum antibody titers of at least of at least 10 minutes are used (see Chapter 11 for more infor-
1:80 as determined with hemagglutination-inhibition correlate mation on disinfection). These disinfectants will also inactivate
well with protection. other enteric viruses. Steam cleaning should be used on surfaces
Concern has been raised that currently available CPV vac- that cannot be otherwise disinfected, and dishwashers that attain
cines may not provide adequate protection against CPV-2c temperatures of at least 75°C should be used to wash dishes. For
infection, because of reports of CPV-2c enteritis in vaccinated shelters, quarantine periods of 2 weeks have been recommended
dogs.51 Furthermore, the serum of animals immunized against before introduction of puppies that might be shedding virus,
CPV-2, CPV-2a, and CPV-2b poorly recognized CPV-2c.52 because shedding rarely continues for longer than 10 days.56 The
However, experimental challenge studies have demonstrated bathing of recovered puppies can help to remove virus that per-
strong protection against CPV-2c challenge when dogs are sists on the haircoat. Rodent and insect vector control can also be
immunized with vaccines that contain CPV-2.53,54 The extent to used to prevent spread of the virus in the environment. Outdoor
which maternal antibody against non-CPV-2c strains protects grassy areas and dirt are difficult or impossible to adequately
against infection with CPV-2c requires investigation. disinfect, and so only immunized animals should be allowed on
Both inactivated and attenuated live vaccines are available these areas. The use of bleach on these areas is not recommended
for prevention or reduction of canine enteric coronavirus infec- because of adverse environmental effects from runoff.
tion, but because the disease is generally mild or inapparent and The prevalence and impact of enteric viral infections can
primarily occurs in puppies younger than 6 weeks of age, their also be reduced through regular removal of fecal contamina-
use has not been generally recommended.55 tion, prolonged exposure of contaminated surfaces to sunlight
and drying, and elimination of stressors such as overcrowding,
Prevention transport, poor nutrition, and concurrent infections such as
intestinal parasites.
Prevention of CPV-2 enteritis requires immunization and appro-
priate quarantine, isolation, cleaning, and disinfection proce- Public Health Aspects
dures. Proper immunization is the most effective method. Puppies
that are incompletely vaccinated should not be introduced into Enteric viral pathogens of dogs, including CPV-2 variants, do
environments where there has been a history of parvoviral enteri- not infect humans. However, other infectious causes of gastro-
tis and adequate environmental disinfection cannot be guaran- enteritis in puppies have the potential to be zoonotic, and co-
teed. Although extremely resistant to disinfectants, parvoviruses infections with these pathogens can occur, so caution should be
can be inactivated with a 1 in 30 dilution of household bleach; maintained when handling puppies with diarrhea. Precautions
potassium peroxymonosulfate (Trifectant, Virkon S); accelerated used in isolation should be sufficient to prevent enteric zoonoses
hydrogen peroxide; or high-level chemical disinfectants such as (see Chapter 11).

CASE EXAMPLE Integument: Full, shiny haircoat. No ectoparasites were seen.


Eyes, ears, nose, and throat: Enophthalmos and a dry nasal
planum were present.
Signalment: “Sam”, an 8-week old intact male Hungarian Musculoskeletal: Body condition score 2/9. The dog was
vizsla puppy from northern California. ambulatory, but emaciated and weak.
History: Sam was acquired 6 days ago from a breeder, and Cardiovascular: Weak but synchronous femoral pulses. No
at that time was eating and active. The next day Sam was murmurs or arrhythmias ausculted.
examined at a local veterinary clinic, and a routine fecal Respiratory: No clinically significant findings.
examination revealed infection with Giardia and Isospora spp. Gastrointestinal: No evidence of abdominal pain on palpation.
Treatment with sulfamethoxazole was initiated, but 1 day Fluid-filled intestinal tract. Full urinary bladder noted.
later the puppy developed diarrhea. The next day, lethargy Rectal examination: Bloody diarrhea was present on the
and vomiting were noted. Vomiting and diarrhea occurred thermometer. Rectal examination was not performed.
every few hours. The diarrhea contained fresh blood and Lymph nodes: All lymph nodes were within normal limits.
the vomitus consisted of clear, frothy fluid. Inappetence and Laboratory Findings:
intermittent vomiting continued for the next 2 days. CBC:
Current Medications: None HCT 31.5% (40-55%)
Other Medical History: Vaccination with an attenuated MCV 62.6 fL (65-75 fL)
live CDV, CPV-2, and canine parainfluenza vaccine was MCHC 35.6 g/dL (33-36 g/dL)
performed when Sam was 6 weeks of age. There were no WBC 530 cells/µL (6000-13,000 cells/µL)
other dogs in the household, only one adult cat. Neutrophils 16 cells/µL (3000-10,500 cells/µL)
Physical Examination: Lymphocytes 504 cells/µL (1000-4000 cells/µL)
Body weight: 4.2 kg Monocytes 11 cells/µL (150-1,200 cells/µL)
General: Quiet, alert, responsive. Estimated to be 7% to Platelets clumped but appeared adequate.
8% dehydrated, T = 103.4°F (39.7°C), HR = 240 beats/min, Serum Chemistry Profile:
RR = 36 breaths/min, mucous membranes pale pink and tacky, Sodium 143 mmol/L (145-154 mmol/L)
CRT = 2 seconds. Potassium 3.8 mmol/L (3.6-5.3 mmol/L)

Continued
150 SECTION 1  Viral Diseases

Chloride 106 mmol/L (108-118 mmol/L) Diagnosis: Enteritis and leukopenia secondary to canine
Bicarbonate 23 mmol/L (16-26 mmol/L) parvoviral infection.
Phosphorus 4.8 mg/dL (3.0-6.2 mg/dL) Treatment: Sam was admitted to isolation and treated
Calcium 10.3 mg/dL (9.7-11.5 mg/dl) aggressively with intravenous lactated Ringer’s solution that
BUN 6 mg/dL (5-21 mg/dL) was supplemented with 20 mEq KCl/L, famotidine (0.5 mg/kg
Creatinine <0.2 mg/dL (0.3-1.2 mg/dL), glucose 133 mg/dL IV q12h), ondansetron (0.5 mg/kg IV q12h), ampicillin (22 mg/
(64-123 mg/dL) kg IV q6h), and enrofloxacin (5 mg/kg IV q24h). Because of
Total protein 4.3 g/dL (5.4-7.6 g/dL) persistent vomiting, maropitant citrate (1 mg/kg IV q24h) was
Albumin 2.4 g/dL (3.0-4.4 g/dL) added as a second antiemetic. Blood glucose concentration
Globulin 1.9 g/dL (1.8-3.9 g/dL) was monitored three times daily, but supplementation with
ALT 22 U/L (19-67 U/L), AST 15 U/L (19-42 U/L) dextrose was not required. Bloody diarrhea that contained
ALP 179 U/L (21-170 U/L) sloughed intestinal mucosa occurred every few hours for
Creatine kinase 274 U/L (51-399 U/L) the first 24 hours. The neutrophil count was 9 cells/µL the
Gamma GT <3 U/L (0-6 U/L) following day, but by day 4 of hospitalization was 261 cells/µL,
Cholesterol 278 mg/dL (135-361 mg/dL) with 241 moderately toxic bands/µL and a lymphocyte count
Total bilirubin 0.1 mg/dL (0-0.2 mg/dL) of 1226 cells/µL. There was gradual clinical improvement
Magnesium 2.0 mg/dL (1.5-2.6 mg/dL). during this time, and Sam began to ingest small quantities
Imaging Findings: of cottage cheese and rice on day 4. He was discharged from
Abdominal radiographs: The stomach wall appeared the hospital on day 5.
thickened. Numerous round gas opacities were present Comments: Despite the negative fecal antigen ELISA assay,
in a loop of intestine in the right cranial abdomen, which CPV enteritis was suspected in this dog based on the history,
were believed to represent gas in the ascending colon clinical signs, and severe leukopenia. The finding of leukopenia
or duodenum. Multiple additional gas opacities were and diarrhea is not diagnostic for parvoviral enteritis, because
present in a loop of intestine in the mid right abdomen at it can also be caused by other severe enteritides such as
the level of the fourth lumbar vertebra. There was reduced salmonellosis. The positive fecal real-time PCR assay for CPV
serosal detail compatible with the age of the animal. supported the diagnosis, but because positive fecal PCR
Changes were considered consistent with the presence of results can occur in healthy dogs with some assays, and also
gastroenterocolitis. after vaccination, the fecal PCR result alone did not confirm the
Microbiologic Testing: diagnosis of CPV enteritis. The diagnosis was therefore made
Fecal Parvovirus Antigen ELISA: Negative on the basis of the combination of findings. The neutropenia
Fecal Enteric Real-time PCR Panel: PCR positive for CPV in in this dog was profound. Infection likely occurred before the
fecal specimen. PCR negative for Clostridium difficile toxin owner acquired the puppy because the diarrhea began only 2
A and toxin B genes, Cryptosporidium spp., Salmonella spp., days after the puppy was purchased.
and Giardia spp. DNA.
Fecal Zinc Sulfate Centrifugal Flotation: Negative for
parasite ova.

SUGGESTED READINGS 6. Pratelli A, Buonavoglia D, Tempesta M, et  al. Fatal canine par-
vovirus type-1 infection in pups from Italy. J Vet Diagn Invest.
Carmichael LE. An annotated historical account of canine parvovirus. 1999;11:365-367.
J Vet Med Ser B. 2005;52(7-8):303-311. 7. Ohshima T, Kawakami K, Abe T, et al. A minute virus of canines
Iris K, Leontides LS, Mylonakis ME, et al. Factors affecting the occur- (MVC: canine bocavirus) isolated from an elderly dog with severe
rence, duration of hospitalization and final outcome in canine parvo- gastroenteritis, and phylogenetic analysis of MVC strains. Vet
virus infection. Res Vet Sci. 2010;89:174-178. Microbiol. 2010;145:334-338.
8. Eminaga S, Palus V, Cherubini GB. Minute virus as a possible cause
REFERENCES of neurological problems in dogs. Vet Rec. 2011;168:111-112.
1. Carmichael LE. An annotated historical account of canine parvovi- 9. Houston DM, Ribble CS, Head LL. Risk factors associated with
rus. J Vet Med Ser B. 2005;52(7-8):303-311. parvovirus enteritis in dogs: 283 cases (1982-1991). J Am Vet Med
2. Hoelzer K, Parrish CR. The emergence of parvoviruses of carni- Assoc. 1996:542-546.
vores. Vet Res. 2010;41:39. 10. Glickman LT, Domanski LM, Patronek GJ, et  al. Breed-related
3. Clegg SR, Coyne KP, Parker J, et al. Molecular epidemiology and risk factors for canine parvovirus enteritis. J Am Vet Med Assoc.
phylogeny reveal complex spatial dynamics in areas where canine 1985;187:589-594.
parvovirus is endemic. J Virol. 2011;85:7892-7899. 11. Kalli I, Leontides LS, Mylonakis ME, et  al. Factors affecting the
4. Ikeda Y, Nakamura K, Miyazawa T, et  al. Feline host range of occurrence, duration of hospitalization and final outcome in canine
canine parvovirus: recent emergence of new antigenic types in cats. parvovirus infection. Res Vet Sci. 2010;89:174-178.
Emerg Infect Dis. 2002;8:341-346. 12. Brady S, Norris JM, Kelman M, et al. Canine parvovirus in Aus-
5. Battilani M, Balboni A, Ustulin M, et al. Genetic complexity and tralia: the role of socio-economic factors in disease clusters. Vet J.
multiple infections with more Parvovirus species in naturally 2012;193:522-528.
infected cats. Vet Res. 2011;42:43.
CHAPTER 14  Canine Parvovirus Infections and Other Viral Enteritides 151

13. Li L, Pesavento PA, Shan T, et al. Viruses in diarrheic dogs include 36. Marulappa SY, Kapil S. Simple tests for rapid detection of canine
novel kobuviruses and sapoviruses. J Gen Virol. 2011;92:2534-2541. parvovirus antigen and canine parvovirus-specific antibodies. Clin
14. Pesavento PA, Li L, Leutenegger C, et-al. Canine circovirus is asso- Vaccine Immunol. 2009;16:127-131.
ciated with vascular damage in dogs. Proceedings of the 2nd sympo- 37. Decaro N, Elia G, Desario C, et al. A minor groove binder probe
sium of the International Society for Companion Animal Infectious real-time PCR assay for discrimination between type 2-based
Diseases. San Francisco, CA. 2012;A01. vaccines and field strains of canine parvovirus. J Virol Methods.
15. Ntafis V, Xylouri E, Radogna A, et al. Outbreak of canine norovi- 2006;136:65-70.
rus infection in young dogs. J Clin Microbiol. 2010;48:2605-2608. 38. Mochizuki M, Ohshima T, Une Y, et al. Recombination between
16. Mesquita JR, Barclay L, Nascimento MS, et al. Novel norovirus in vaccine and field strains of canine parvovirus is revealed by iso-
dogs with diarrhea. Emerg Infect Dis. 2010;16:980-982. lation of virus in canine and feline cell cultures. J Vet Med Sci.
17. Martella V, Lorusso E, Decaro N, et  al. Detection and molecu- 2008;70:1305-1314.
lar characterization of a canine norovirus. Emerg Infect Dis. 39. Nho WG, Sur JH, Doster AR, et al. Detection of canine parvovirus
2008;14:1306-1308. in naturally infected dogs with enteritis and myocarditis by in situ
18. Evermann JF, Abbott JR, Han S. Canine coronavirus-associated hybridization. J Vet Diagn Invest. 1997;9:255-260.
puppy mortality without evidence of concurrent canine parvovirus 40. Waldvogel AS, Hassam S, Stoerckle N, et  al. Specific diagnosis
infection. J Vet Diagn Invest. 2005;17:610-614. of parvovirus enteritis in dogs and cats by in situ hybridization.
19. Macartney L, McCandlish IA, Thompson H, et al. Canine parvovi- J Comp Pathol. 1992;107:141-146.
rus enteritis 2: pathogenesis. Vet Rec. 1984;115:453-460. 41. Groeneveld AB, Navickis RJ, Wilkes MM. Update on the compara-
20. Otto CM, Rieser TM, Brooks MB, et  al. Evidence of hypercoag- tive safety of colloids: a systematic review of clinical studies. Ann
ulability in dogs with parvoviral enteritis. J Am Vet Med Assoc. Surg. 2011;253:470-483.
2000;217:1500-1504. 42. Mohr AJ, Leisewitz AL, Jacobson LS, et al. Effect of early enteral
21. Hayes MA, Russell RG, Babiuk LA. Sudden death in young dogs nutrition on intestinal permeability, intestinal protein loss, and out-
with myocarditis caused by parvovirus. J Am Vet Med Assoc. come in dogs with severe parvoviral enteritis. J Vet Intern Med.
1979;174:1197-1203. 2003;17:791-798.
22. Lenghaus C, Studdert MJ, Finnie JW. Acute and chronic canine 43. Duffy A, Dow S, Ogilvie G, et  al. Hematologic improvement in
parvovirus myocarditis following intrauterine inoculation. Aust dogs with parvovirus infection treated with recombinant canine
Vet J. 1980;56:465-468. granulocyte-colony stimulating factor. J Vet Pharmacol Ther.
23. Bastianello SS. Canine parvovirus myocarditis: clinical signs and 2010;33:352-356.
pathological lesions encountered in natural cases. J S Afr Vet Assoc. 44. Savigny MR, Macintire DK. Use of oseltamivir in the treatment of
1981;52:105-108. canine parvoviral enteritis. J Vet Emerg Crit Care (San Antonio).
24. Schatzberg SJ, Haley NJ, Barr SC, et  al. Polymerase chain reac- 2010;20:132-142.
tion (PCR) amplification of parvoviral DNA from the brains 45. Otto CM, Jackson CB, Rogell EJ, et al. Recombinant bactericidal/
of dogs and cats with cerebellar hypoplasia. J Vet Intern Med. permeability-increasing protein (rBPI21) for treatment of parvovirus
2003;17:538-544. enteritis: a randomized, double-blinded, placebo-controlled trial. J Vet
25. Lenghaus C, Studdert MJ. Generalized parvovirus disease in neona- Intern Med. 2001;15:355-360.
tal pups. J Am Vet Med Assoc. 1982;181:41-45. 46. Otto CM, Drobatz KJ, Soter C. Endotoxemia and tumor necrosis
26. Decaro N, Martella V, Elia G, et al. Tissue distribution of the anti- factor activity in dogs with naturally occurring parvoviral enteritis. J
genic variants of canine parvovirus type 2 in dogs. Vet Microbiol. Vet Intern Med. 1997;11:65-70.
2007;121:39-44. 47. Ling M, Norris JM, Kelman M, et al. Risk factors for death from
27. Schaudien D, Polizopoulou Z, Koutinas A, et al. Leukoencephalop- canine parvoviral-related disease in Australia. Vet Microbiol.
athy associated with parvovirus infection in Cretan hound puppies. 2012;158:280-290.
J Clin Microbiol. 2010;48:3169-3175. 48. Goddard A, Leisewitz AL, Christopher MM, et al. Prognostic use-
28. Agungpriyono DR, Uchida K, Tabaru H, et  al. Subacute mas- fulness of blood leukocyte changes in canine parvoviral enteritis.
sive necrotizing myocarditis by canine parvovirus type 2 infec- J Vet Intern Med. 2008;22:309-316.
tion with diffuse leukoencephalomalacia in a puppy. Vet Pathol. 49. Mason MJ, Gillett NA, Muggenburg BA. Clinical, pathological,
1999;36:77-80. and epidemiological aspects of canine parvoviral enteritis in an
29. Iris K, Leontides LS, Mylonakis ME, et  al. Factors affecting the unvaccinated closed beagle colony: 1978-1985. J Am Anim Hosp
occurrence, duration of hospitalization and final outcome in canine Assoc. 1987;23:183-192.
parvovirus infection. Res Vet Sci. 2010;89:174-178. 50. Horner GW. Canine parvovirus in New Zealand: epidemiological
30. Woldemeskel M, Liggett A, Ilha M, et  al. Canine parvovirus-2b- features and diagnostic methods. N Z Vet J. 1983;31:164-166.
associated erythema multiforme in a litter of English Setter dogs. 51. Decaro N, Desario C, Elia G, et al. Evidence for immunisation fail-
J Vet Diagn Invest. 2011;23:576-580. ure in vaccinated adult dogs infected with canine parvovirus type
31. Favrot C, Olivry T, Dunston SM, et  al. Parvovirus infection of 2c. New Microbiol. 2008;31:125-130.
keratinocytes as a cause of canine erythema multiforme. Vet Pathol. 52. Cavalli A, Martella V, Desario C, et al. Evaluation of the antigenic
2000;37:647-649. relationships among canine parvovirus type 2 variants. Clin Vaccine
32. Decaro N, Desario C, Beall MJ, et al. Detection of canine parvovi- Immunol. 2008;15:534-539.
rus type 2c by a commercially available in-house rapid test. Vet J. 53. Siedek EM, Schmidt H, Sture GH, et al. Vaccination with canine
2010;184:373-375. parvovirus type 2 (CPV-2) protects against challenge with viru-
33. Schmitz S, Coenen C, Konig M, et al. Comparison of three rapid lent CPV-2b and CPV-2c. Berl Munch Tierarztl Wochenschr.
commercial canine parvovirus antigen detection tests with electron 2011;124:58-64.
microscopy and polymerase chain reaction. J Vet Diagn Invest. 54. Larson LJ, Schultz RD. Do two current canine parvovirus type 2
2009;21:344-345. and 2b vaccines provide protection against the new type 2c variant?
34. Greene CE, Decaro N. Canine viral enteritis. In: Greene CE, ed. Vet Ther. 2008;9:94-101.
Infectious Diseases of the Dog and Cat. 4th ed. St Louis, MO: Else- 55. Welborn LV, DeVries JG, Ford R, et al. 2011 AAHA canine vac-
vier Saunders; 2012. cination guidelines. J Am Anim Hosp Assoc. 2011;47:1-42.
35. Patterson EV, Reese MJ, Tucker SJ, et  al. Effect of vaccination 56. Appel LD, Barr SC. Canine parvovirus and coronavirus. In: Miller
on parvovirus antigen testing in kittens. J Am Vet Med Assoc. L, Hurley KF, eds. Infectious Disease Management in Animal Shel-
2007;230:359-363. ters. Ames, IA: Wiley-Blackwell; 2009:197-208.
CHAPTER 15

Canine Distemper Virus Infection


Jane E. Sykes

Ferrets also are highly susceptible to distemper, but domestic


Overview of Canine Distemper cats are not affected. In addition, distemper occurs in a variety
First Described: 1905, Henri Carré, France1 of wildlife species that belong to the families Canidae, Musteli-
dae, Procyonidae, and Felidae (see Overview). Virus shed by
Cause: Canine distemper virus (family Paramyxoviridae, sub-
wildlife species such as raccoons can infect domestic dogs, but
family Paramyxovirinae, genus Morbillivirus)
virus that is shed by dogs may also be a significant threat to
Affected Hosts: Dogs and other Canidae such as coyotes, wildlife populations, with catastrophic outbreaks of disease and
foxes, and wolves; Procyonidae (raccoons, pandas); Mus- high mortality rates. In domestic dogs with signs of infectious
telidae (ferrets, mink, skunks, otters). Large wild Felidae upper respiratory disease (“kennel cough”), distemper is an
can also be affected. important differential diagnosis, and distemper can also mimic
Geographic Distribution: Worldwide canine parvovirus (CPV) enteritis. In fact, dual infections with
CDV and CPV are probably underrecognized.
Mode of Transmission: Oronasal contact with virus in secre- Worldwide, at least eight different geographic lineages
tions or excretions, droplet nuclei, and large particle aero- (also referred to as genotypes or clades) of CDV exist based
sol transmission on sequence analysis of the H gene: Asia-1, Asia-2, America-1,
Major Clinical Signs: Fever, lethargy, inappetence, vomit- America-2, Europe-1/South America-1, Europe-2 (European
ing, diarrhea, dehydration, tachypnea, cough, conjunc- wildlife), Europe-3 (Arctic-like), and South Africa. In addition,
tivitis, neurologic signs, footpad, and nasal planum strains recognized in Argentina, Africa, Asia, and Mexico may
hyperkeratosis represent separate geographic lineages.1,2 In the United States,
Differential Diagnoses: Canine parvovirus infection, other vaccine strains belong to the America-1 lineage, whereas most
infectious causes of gastroenteritis, other causes of field strains belong to the America-2 lineage. Strains that belong
canine infectious respiratory disease, rabies, parasitic gas- to the Europe-2 and Europe-3 lineages have also been detected
trointestinal and respiratory disease, toxins such as lead in North American dogs.3
and ethylene glycol, gastrointestinal foreign body, dietary Although vaccination has reduced the incidence of the dis-
indiscretion, protozoal meningoencephalitis, systemic ease, distemper still remains important where large numbers of
fungal infections such as cryptococcosis, portosystemic
shunting with hepatic encephalopathy
Human Health Significance: CDV has been used as a model to Fusion protein
study measles virus infection. There is some evidence that
Hemagglution
it may be involved in the pathogenesis of Paget’s disease,
a chronic, focal skeletal disorder of the elderly. SH protein

Etiology and Epidemiology


Canine distemper is an important disease of domestic dogs
and wild animals worldwide. It is caused by canine distem-
per virus (CDV), an enveloped, pleomorphic RNA virus that Matrix protein
belongs to the genus Morbillivirus (family Paramyxoviridae).
CDV is closely related to human measles virus and rinderpest Nucleoprotein
virus and has been used as a model to study the pathogenesis of
Polymerase
measles virus infections. The outer envelope of CDV contains
hemagglutinin (H) and fusion (F) proteins, which are impor-
tant in cellular attachment and entry (Figure 15-1). Infection of
dogs can lead to a severe, multisystemic disease that primarily FIGURE 15-1  Structure of CDV. The outer envelope contains hemagglutinin (H) and
affects the gastrointestinal, respiratory, and neurologic systems fusion (F) proteins, which are connected to the nucleocapsid by a matrix (M) protein, which
(Figure 15-2). is the most abundant protein in the virion.

152
CHAPTER 15  Canine Distemper Virus Infection 153

FIGURE 15-2  Anatomic sites targeted by CDV infection.

young dogs with inadequate immunity are housed together, such


as in kennels, large breeding facilities, and shelter environments.
Puppies that have lost their maternal antibody are predisposed
in regions where vaccination is performed, but occasionally dis-
ease occurs in older, vaccinated dogs.4-6 Distemper also occurs
in regions where vaccination of dogs is not performed or is
poorly timed. As an enveloped virus, CDV is susceptible in the
environment (surviving less than 1 day at room temperature)
and is readily inactivated by heat, drying, and disinfectants;
thus, contact between infected dogs is important to maintain
transmission of the virus.

Clinical Features
Signs and Their Pathogenesis
CDV is highly contagious and is spread through droplet nuclei and
large-particle aerosol transmission. Dogs are generally exposed
to CDV through contact with infected oronasal secretions, which FIGURE 15-3  Lymph node histopathology of a 5-month-old German shepherd
may be shed by subclinically or clinically affected dogs. The virus mix with distemper. The dog had been adopted from a shelter and subsequently devel-
initially infects monocytes within lymphoid tissue in the upper oped neurologic signs, diarrhea, and pneumonia. There is marked lymphoid necrosis.
respiratory tract and tonsils and is subsequently disseminated via H&E stain.
the lymphatics and blood to the entire reticuloendothelial sys-
tem. The viral hemagglutinin binds to a molecule on the sur-
face of host cells known as the signaling lymphocytic activation (8 to 9 days after infection), after which CDV infects cells of the
molecule (SLAM, also known in humans as CD150). SLAM is a respiratory, gastrointestinal tract, central nervous system (CNS),
membrane glycoprotein of the immunoglobulin superfamily. It is urinary tract, and skin, as well as red and white blood cells,
important for cell entry by CDV as well as other morbilliviruses including additional lymphoid cells. In this stage of infection,
and is a key molecule in the pathogenesis of disease.7 SLAM is CDV infects a variety of cell lineages, including epithelial, mes-
expressed by immature thymocytes, activated lymphocytes, mac- enchymal, neuroendocrine, and hematopoietic cells, and forms
rophages, and dendritic cells. Direct viral destruction of a sig- intracytoplasmic but also intranuclear inclusions (Figure 15-4).
nificant proportion of the lymphocyte population, and especially Infection of epithelial cells by the virus is inefficient and occurs
CD4+ T cells, occurs within the blood, tonsils, thymus, spleen, relatively late in the course of infection, through a SLAM-inde-
lymph nodes, bone marrow, mucosa-associated lymphoid tissue, pendent mechanism that appears to involve the nectin 4 receptor,
and hepatic Kupffer cells (Figure 15-3).8-10 Massive destruction as is true for measles virus.11 CDV is shed in all secretions and
of lymphocytes results in an initial lymphopenia and transient excretions from as early as day 5 after infection, before the onset
fever, which occurs a few days after infection. Subsequently, of clinical signs. Shedding of virus can continue for as long as
there is a second stage of cell-associated viremia and fever 3 to 4 months, but usually resolves after 1 to 2 weeks.
154 SECTION 1  Viral Diseases

A B
FIGURE 15-4  A, Eosinophilic intracytoplasmic and intranuclear viral inclusions (arrows) in the lung of the dog from Figure 15-3. Hematoxylin and eosin stain, 1000× oil magnification.
B, Intracytoplasmic inclusions (arrows) in a circulating leukocyte of a 4-month-old Border collie mix with distemper.

The clinical signs of distemper in dogs vary dramatically


and are highly dependent on virus strain and the age and
immune status of the host, as well as concurrent infections
with other viruses and bacteria. Many dogs experience sub-
clinical infection, whereas others experience rapidly progres-
sive infection followed by death. The incubation period ranges
from 3 to 6 days. Respiratory or gastrointestinal tract signs
may be indistinguishable from those caused by other respira-
tory or enteric viruses and bacteria, or signs may be so mild
that they go unnoticed by the owner. Dogs with respiratory
involvement may exhibit fever, bilateral serous and nasal ocu-
lar discharges, conjunctivitis, and a nonproductive cough.
Secondary bacterial infection, which occurs unhindered as a
result of virus-induced immunosuppression, can lead to the
development of mucopurulent nasal and ocular discharges FIGURE 15-5  Enamel hypoplasia induced by CDV infection.
and bacterial bronchopneumonia, with tachypnea, productive
cough, lethargy, and decreased appetite. Viral destruction of
the gastrointestinal tract epithelium can result in inappetence, sclerosis because of the resemblance of demyelinating CDV
vomiting, diarrhea, electrolyte abnormalities, and dehydra- encephalitis to the pathology of this disease.15 Virus has been
tion. Dogs that mount an intermediate or delayed immune detected within the brains of these dogs with immunohisto-
response may recover from acute illness but fail to elimi- chemistry and reverse-transcriptase PCR assays.16 Persistent
nate the virus completely, which leads to a spectrum of more infection of astrocytes in the CNS by some CDV strains has
chronic disease manifestations, which often involve the uvea, been postulated as a mechanism for this chronic manifesta-
lymphoid organs, footpads, and especially the CNS. Opportu- tion of infection, with transmission from cell to cell through
nistic infections can also develop in these dogs. induction of cell-cell fusion and resultant evasion of the host
Up to 30% of infected dogs develop CNS signs. These usu- immune response.17
ally occur 1 to 6 weeks after the onset of acute illness but can Ocular signs of persistent CDV infection include uveitis, cho-
occur even when initial infection is subclinical. Neurologic rioretinitis, keratoconjunctivitis sicca (KCS), keratitis, and optic
signs are more likely to develop following infection with cer- neuritis, which may be associated with blindness. KCS is thought
tain CDV strains, such as the Snyder Hill strain.12 The types to result from damage to the lacrimal gland by CDV.18 It may
of brain cells that are infected differ with the strain of CDV be transient or permanent, and can lead to keratitis and corneal
involved,13 but neuronal necrosis and atrophy can occur.14 ulceration. The virus itself can also infect the corneal epithelial
Neurologic signs are often progressive and generally do not and stromal cells. Enamel and dentin hypoplasia, manifested as
resolve, so dogs that recover often have residual neurologic irregularities of the teeth, is an incidental finding in recovered
deficits. “Old dog encephalitis” is a poorly characterized, dogs due to infection of the stratum intermedium of developing
progressive immune-mediated demyelinating leukoencepha- teeth (Figure 15-5). Retention and partial eruption of teeth has
lomyelitis induced by CDV that occurs weeks to years after also been reported as a sequela of CDV infection.19,20 Infection
recovery from the acute infection and is difficult to diagnose. of the skin can lead to a cutaneous measles-like rash, although
The term “old dog encephalitis” is a misnomer, because this is uncommonly recognized. Persistent infection of the foot-
affected dogs are not necessarily old. Both CDV and measles pad and nasal planum epithelium leads to hyperkeratosis in
virus have been incriminated as possible agents of multiple these regions (Figure 15-6).
CHAPTER 15  Canine Distemper Virus Infection 155

A B

C
FIGURE 15-6  Nasal planum (A) and footpad (B) hyperkeratosis induced by CDV infection in a 4-month-old Border collie mix with conjunctivitis, mucopurulent nasal and ocular
discharge, and myoclonus. Blepharospasm is also present. C, Histopathology of the footpad of a 1-year old German shepherd mix with distemper. Marked hyperkeratosis is evident. H&E
stain, 40× magnification.

Immunosuppression results from lymphopenia, necrosis undergo degeneration and necrosis, and metaphyseal osteoscle-
of hematopoietic cells in the bone marrow, and dendritic cell rosis occurs (Figure 15-7). This may lead to pain and lameness.
malfunction. Lymphocyte apoptosis also occurs independent CDV RNA has also been detected in bone cells of dogs with
of viral infection of lymphocytes.21 The viral V protein allows hypertrophic osteodystrophy, and CDV may play a role in this
CDV to replicate rapidly in T cells and is critical in CDV- disease.30
mediated immunosuppression. This protein almost completely Finally, transplacental infections with CDV may be associ-
antagonizes IFN-α, TNF-α, Il-6, IFN-γ, and Il-2 in the acute ated with infertility, stillbirth, or abortion and with neurologic
phase of infection.10 Finally, the nucleocapsid (N) protein of signs in puppies that are less than 4 to 6 weeks of age.
morbilliviruses binds to the CD32 (Fc-γ) receptor on B cells,
which results in impaired differentiation of B cells into plasma Physical Examination Findings
cells.22 When the virus binds this receptor on dendritic cells, Physical examination findings in dogs with distemper depend
impaired antigen presentation by dendritic cells results, which on the severity and chronicity of the disease, but include fever
disrupts T-cell function. The most common secondary infections (occasionally up to 106°F or 41.1°C), conjunctivitis, serous to
in distemper are secondary bacterial infections that contribute mucopurulent ocular and nasal discharges, blepharospasm and
to bronchopneumonia. Bordetella bronchiseptica is a common photophobia, tonsillar enlargement and hyperemia, tachypnea,
copathogen. Dogs may be diagnosed with bordetellosis in the and increased lung sounds (see Figure 15-6). Cough may occur
early stages of distemper, and the underlying CDV infection during the examination. Dehydration may also be present, espe-
may be overlooked. Other opportunistic infections that have cially in dogs with gastrointestinal and severe neurologic signs.
been identified in dogs with distemper include toxoplasmosis,23 Rarely, a measles-like cutaneous rash or pustular dermatitis is
salmonellosis,24 nocardiosis,25,26 and generalized demodecosis. evident.
Infection with Pneumocystis carinii was associated with CDV Dogs with chronic disease can be thin or emaciated and
infection in a mink,27 and concurrent neosporosis and canine show hyperkeratosis of the nasal planum and footpads. Puppies
distemper was reported in a raccoon.28 with hyperkeratosis, also known as “hardpad,” have thickened,
Uncommonly, CDV infection has been associated with crusty footpads that resemble those of adult dogs (see Figure
metaphyseal bone lesions in young dogs as a result of infec- 15-6). Myoclonus (involuntary twitching of isolated muscle
tion of osteoclasts, osteoblasts, and osteocytes.29 Infected cells groups) is common and, when mild, is most readily detected
156 SECTION 1  Viral Diseases

different anatomic locations, such as various combinations of


blood, urine, conjunctival smears, and CSF. In dogs with respi-
ratory signs, the presence of conjunctivitis, blepharospasm, and
ocular discharge should raise suspicion for distemper. Distem-
per should always be considered as a diagnosis in young dogs
with neurologic signs, whether or not respiratory or gastroin-
testinal signs are present. Finally, clinicians should maintain a
high suspicion for the presence of concurrent infections such
as parvoviral enteritis, concurrent respiratory viral infection, or
bordetellosis.

Laboratory Abnormalities
Complete Blood Count
The CBC in dogs with acute distemper most commonly reveals
mild anemia and lymphopenia (Table 15-1). However, the lym-
phocyte count may also be normal, especially with more chronic
distemper. Neutropenia, monocytopenia, and thrombocytope-
FIGURE 15-7  Severe, neutrophilic osteomyelitis with submetaphyseal osteone- nia can occur, and occasionally these are severe. Neutrophilia
crosis and extensive bony resorption in a 4-month-old intact male Labrador retriever with a left shift and toxic neutrophils may be present. Rarely,
mix with distemper. The dog was seen for lethargy, mild diarrhea, fever, and reluc- examination of Romanowsky-stained blood smears using light
tance to stand; joint pain was detected on physical examination. Direct IFA on urine microscopy reveals CDV inclusions in the cytoplasm of circulat-
sediment was positive for CDV, but a conjunctival scrape tested negative with direct ing erythrocytes and leukocytes (see Figure 15-4, B).
IFA. Necropsy also revealed pneumonia with intracytoplasmic and intranuclear viral
inclusions. H&E stain.
Serum Biochemical Tests
Abnormalities on the serum biochemistry panel in dogs with
when affected puppies are at rest. Other neurologic signs distemper are nonspecific and include electrolyte changes that
include obtundation, seizures, tremors, opisthotonos, tetrapa- occur with vomiting and diarrhea such as hyponatremia, hypo-
resis, paraparesis, delayed placing reactions, ataxia, and, less kalemia, and hypochloridemia (Table 15-2). Hypoalbuminemia
commonly, behavioral abnormalities, compulsive pacing, and and associated hypocalcemia are also common. Mild increases
vestibular signs such as a head tilt, nystagmus, strabismus, and in liver enzyme activities occur in some dogs, which may reflect
circling.4 Focal seizures may be localized to the head and jaw, hypoxia or secondary infections that occur as a result of trans-
with accompanying foamy hypersalivation (“chewing gum” location of intestinal bacteria.
seizures). Vocalization and apparent blindness can also occur.
In addition to conjunctivitis and ocular discharge, an oph- Urinalysis
thalmologic examination may reveal corneal edema, corneal No specific urinalysis findings are reported in dogs with
ulceration, aqueous flare, chorioretinitis, and uveitis, or optic distemper.
neuritis. Markedly decreased Schirmer tear test results and posi-
tive fluorescein tests may be present.18 Optic neuritis appears as CSF Analysis
a large, pale, fluffy, optic disc with no physiologic depression. CSF analysis may show no clinically significant findings in dogs
Rarely, lameness may be observed. with distemper, especially when chronic demyelinating enceph-
Dogs that have recovered from CDV infection may have alitis is present. In some dogs with CNS involvement, a mild
evidence of dental abnormalities and enamel hypoplasia (see to moderate increase in CSF protein concentration is present
Figure 15-5); healed chorioretinitis lesions, which appear as (usually <100 mg/dL), and there is lymphocytic or monocytic
hyperreflective circular lesions (“gold medallion lesions”); kera- pleocytosis, sometimes with reactivity. Nucleated cell counts are
toconjunctivitis sicca; or residual neurologic signs, especially usually less than 50 cells/µL and rarely less than 100 cells/µL.4
myoclonus.
Diagnostic Imaging
Diagnosis Plain Radiography
When present, abnormalities on thoracic radiography in dogs
Establishment of a diagnosis of distemper is most easily accom- with distemper include pulmonary interstitial to alveolar infil-
plished in dogs that have the full spectrum of clinical abnormali- trates and consolidation compatible with bronchopneumonia,
ties, and especially when myoclonus is present, because there especially in the cranial and ventral lung lobes.
are few other diseases that cause this array of clinical signs.
In dogs with isolated respiratory, gastrointestinal, or neuro- Magnetic Resonance Imaging
logic signs, antemortem diagnosis is more challenging because Dogs with distemper encephalomyelitis can have normal mag-
these signs are less specific, laboratory diagnostic assays can be netic resonance imaging findings. Hyperintense lesions and loss
insensitive, and the presence of attenuated live vaccine virus of contrast between gray and white matter in the cerebellum and/
can lead to false-positive results. A combination of laboratory or the brainstem have been described in T2-weighted images,
tests may be required to establish the diagnosis, and the finding which correspond to demyelination on histopathology.31 Multi-
of a negative result for any assay does not rule out distemper. focal T2 and fluid-attenuated inversion recovery (FLAIR) hyper-
Sensitivity for detection of CDV using assays that detect virus intensities can occur in other locations in the brain, and there
may also be increased by testing of multiple specimens from may be mild meningeal contrast enhancement.
CHAPTER 15  Canine Distemper Virus Infection 157

TABLE 15-1
Complete Blood Count Findings at Admission in 50 Dogs Diagnosed with Distemper at the UC Davis VMTH
Percent below Percent within Percent above Range for
Reference the Reference the Reference the Reference Dogs with
Test Range Range Range Range Distemper
Hematocrit (%) 40-55 80 20 0 17-53
MCV (fL) 65-75 26 72 2 58-80
MCHC (g/dL) 33-36 10 74 16 28-37
Neutrophils (cells/µL) 3000-10,500 12 32 56 16-27,765
Band neutrophils 0-rare 0 72 28 0-4442
(cells/µL)
Monocytes (cells/µL) 150-1200 4 62 34 24-5202
Lymphocytes (cells/µL) 1000-4000 54 42 4 110-5575
Eosinophils (cells/µL) 0-1500 0 100 0 0-1114
Platelets (cells/µL) 150,000-400,000 14 66 20 17,000-594,000

Note: Adult reference ranges were used.

TABLE 15-2
Findings on Serum Biochemistry Analysis in 27 Dogs with Distemper at the UC Davis VMTH
Percent below Percent within Percent above Range for
Reference the Reference the Reference the Reference Dogs with Number of
Test Range Range Range Range Distemper Dogs Tested
Sodium (mmol/L) 145-154 56 40 5 106-158 26
Potassium (mmol/L) 3.6-5.3 12 79 9 2.8-5.9 26
Chloride (mmol/L) 108-118 40 56 5 65-130 26
Bicarbonate (mmol/L) 16-26 0 86 14 16-34 26
Calcium (mg/dL) 9.7-11.5 25 68 7 5.1-12.7 27
Phosphorus (mg/dL) 3.0-6.2 7 50 43 2.6-10.0 27
Creatinine (mg/dL) 0.3-1.2 11 89 0 0-1.2 27
BUN (mg/dL) 5-21 7 91 2 0.6-25 27
Albumin (g/dL) 3.0-4.4 68 32 0 0.9-3.7 27
Globulin (g/dL) 1.8-3.9 7 82 11 1.5-5.3 27
Cholesterol (mg/dL) 135-361 9 86 5 88-385 26
Total bilirubin (mg/dL) 0-0.2 0 80 20 0-0.8 26
ALT (U/L) 19-67 7 75 18 8-389 26
ALP (U/L) 21-170 0 77 23 28-322 26

Note: Adult reference ranges were used by the laboratory.

Microbiologic Tests distinguished from vaccine virus in dogs that have been recently
Microbiological tests for CDV infection are shown in Table 15-3. vaccinated with attenuated live vaccines.

Virus Isolation Cytologic Demonstration of CDV Inclusions


CDV can be readily isolated in SLAM transfected Vero cell Intracytoplasmic viral inclusions can occasionally be seen in
lines, which show cytopathic effects within 24 hours of inocula- circulating blood cells of affected dogs, especially early in
tion. Isolation is generally used as a research tool rather than infection (see Figure 15-4, B). Inclusions may also be visible
for commercial diagnostic purposes. Wild-type virus must be in the cytoplasm of conjunctival epithelial cells. Epithelial
158 SECTION 1  Viral Diseases

cells can be scraped from the conjunctiva using a curette do not distinguish between vaccine and wild-type strains. Assays
after application of a topical ophthalmic local anesthetic have been developed that differentiate between field and vaccine
preparation. The cells are smeared onto a glass slide, stained strains of CDV.36 False-positive test results should not occur
with a Romanowsky stain and examined with light micros- after use of recombinant CDV vaccines. More research is needed
copy. Unfortunately, this has low sensitivity for diagnosis of to clarify the influence of vaccination on the results of RT-PCR
distemper. for distemper. Quantitation of RT-PCR results may ultimately
be helpful in differentiating vaccination with attenuated virus
Immunostaining for CDV Antigen ­vaccines (low viral load) and natural infection (high viral load).
Direct immunofluorescent antibody (IFA) techniques are often False-negative RT-PCR results can occur if the virus is pres-
used in practice to detect CDV antigen in a variety of cell types, ent in low levels at the site from which specimens are collected.
including conjunctival epithelial cells, leukocytes in a buffy coat Because RNA is labile, degradation of viral RNA during speci-
smear, urine sediment, or tissues obtained at necropsy. Immu- men collection and transport to the laboratory can also lead to
noperoxidase antibody can also be used. Immunostaining is false-negative PCR results. In outbreak situations, collection of
more sensitive than identification of CDV inclusions with rou- specimens from multiple dogs (at least 5 to 10) may increase the
tine stains, but false negatives still occur when low quantities of chance of positive test results.37 The diagnosis is strengthened
virus are present in the specimen. when positive PCR assay results are combined with positive
Transient false positive results might occur after vaccination results of other assays such as direct IFA.
with attenuated live vaccines, so the results must be interpreted
with caution in recently vaccinated dogs. False positives also Serology
have the potential to occur if nonspecific fluorescence is inter- Antibodies to CDV can be measured in the laboratory using
preted as a positive result by inexperienced laboratory techni- serum neutralization (SN) assays (see Chapter 2). ELISA assays
cians. Further research is required to understand the prevalence that detect IgG and IgM are also available for routine diagnosis
of, and influence of vaccination on false positive direct IFA of distemper in dogs, which have improved sensitivity and speci-
results for diagnosis of distemper. ficity over SN.38 In one study, IgM antibodies appeared 1 week
before SN titers.39 Use of serology for diagnosis of distemper is
Antigen Detection ELISA Assays complicated by the confounding effect of vaccination. This is
ELISA assays that detect CDV antigen in serum (Table 15-3) are especially true in shelter environments where all dogs are vacci-
available commercially as in-practice kits in some countries. The nated on intake. Nevertheless, a fourfold rise in titer over a 2- to
sensitivity and specificity of these assays has not been well stud- 4-week period supports recent infection. Acute and convalescent
ied, and there are no published reports in the scientific literature sera should be submitted to the same laboratory, because inter-
of the performance of commercially available assays, only those laboratory variation in assay results occurs. Ideally, an aliquot
in the research and development phase. One assay had a low of the acute serum specimen should be retested at the same time
sensitivity in dogs that were naturally infected with distemper, the convalescent titer is determined. Antibodies within the CSF
with positive results in only 27% of 26 dogs at initial exami- can be quantified and compared to those in serum to determine
nation and 12% when the dogs were examined 2 to 3 weeks whether local production of antibody in the CSF is likely. Dogs
later.32 False-positive results occurred for up to 4 weeks after with delayed-onset CDV encephalomyelitis have high levels of
vaccination in 20% of 40 dogs immunized with attenuated live antibody in the CSF when compared with serum.40
viral vaccines. Another lateral flow (immunochromatography) The results of serologic tests can be used to assess the need
assay had sensitivity equivalent to that of an RT-PCR assay for vaccination, and also to determine which dogs are protected
when applied to conjunctival swab specimens, but sensitivity and therefore of low risk for development of disease and to some
was 90% and 86% when the assay was applied to blood lym- extent, virus shedding in outbreak situations. Serum neutraliza-
phocytes and nasal swab specimens from dogs with distemper.33 tion titers of at least 1:16 to 1:20 correlate with protection after
vaccination. Titers of 1:100 or more correlate with protection in
Molecular Diagnosis Using the Polymerase Chain Reaction puppies that have received maternal antibodies.41 Because of the
Reverse transcriptase-PCR (RT-PCR) assays are offered by a presence of cell-mediated immunity, negative serologic results
number of commercial veterinary diagnostic laboratories for do not imply lack of protection. Point-of-care ELISA assays are
diagnosis of distemper, sometimes as part of a panel that includes also available for semiquantitative measurement of antibodies
assays for other canine respiratory pathogens (see Chapter 17). to CDV. One assay (Synbiotics TiterCHEK CDV/CPV) had a
Most assays detect a portion of the N protein gene. RT-PCR sensitivity of 76% and a specificity of 92% compared with SN
can be performed on whole blood, buffy coat, skin biopsies, when performed as a point-of-care assay.42 The low sensitivity
conjunctival scrapings, urine, CSF, transtracheal washes, nasal would tend to overestimate the need for vaccination, which is
and oropharyngeal swabs, and a variety of other tissues collected preferable to an assay with low specificity, which might result
at necropsy (e.g., lung, small intestine, stomach, kidney, brain, in unnecessary exposure of a susceptible dog to infected dogs.
bladder, lymphoid tissues). In one study of a limited number of
experimentally infected dogs, conjunctival swabs were superior Pathologic Findings
to whole blood, nasal flushes, and urine for PCR diagnosis of Gross Pathologic Findings
CDV infection,34 whereas another study of naturally infected In addition to physical examination abnormalities described ear-
dogs showed highest viral loads in blood, followed by conjunc- lier, gross pathologic findings in canine distemper include thymic
tival swabs, and then urine.35 RT-PCR is more sensitive than atrophy, pulmonary congestion and consolidation, liquid intesti-
direct IFA, but false positives may be more likely to occur after nal contents, and lymph node congestion and enlargement. Less
vaccination with attenuated live virus vaccines, because RT-PCR common findings are mild pleural, pericardial, and/or peritoneal
assays that are currently available for routine diagnostic purposes effusion and, uncommonly, visceral congestion and ecchymotic
CHAPTER 15  Canine Distemper Virus Infection 159

TABLE 15-3
Diagnostic Assays Available for Distemper in Dogs
Assay Specimen Type Target Performance
Virus isolation Conjunctival and nasal CDV False negatives can occur in specimens
swabs, blood, urine, trans- that contain low numbers of virus
tracheal wash specimens, particles. Attenuated live vaccine
necropsy specimens virus may also be isolated. Not
widely offered for routine diagnostic
purposes.
Direct immunostaining Smears made from con- CDV antigen False negatives can occur in specimens
(such as direct fluorescent junctival scrapings, urine that contain low numbers of virus
antibody) sediment, respiratory lavage particles. False positives can occur if
specimens, impression nonspecific fluorescence is incorrectly
smears of tissue specimens interpreted as a positive result. Pos-
sibility of false positives after recent
vaccination with attenuated live CDV
vaccines.
Distemper ELISA antigen See Virus isolation CDV antigen Inexpensive and rapid. Sensitivity and
assay specificity in naturally infected dogs
have not been well established. False
positives can occur for weeks after
vaccination with attenuated live
CDV vaccines.
RT-PCR See Virus isolation CDV RNA Sensitivity and specificity can vary
depending on assay design. Testing
specimens from multiple anatomic
sites or combining PCR assays with
other diagnostic tests for CDV
improves sensitivity. Attenuated live
vaccine virus may be detected for days
to weeks after vaccination. Because
of the high sensitivity of some assays,
the significance of a positive result in
relation to disease may be difficult to
interpret. False-negative results can
occur when virus levels are low or as
a result of degradation of viral nucleic
acid during specimen transport.
Serology (IgG and IgM Serum and/or CSF Antibodies against Acute and convalescent sera are
ELISA; SN) CDV antigens required for diagnosis of acute dis-
temper, so diagnosis is retrospective.
False positives can occur with recent
CDV vaccination.
Histopathology Usually necropsy specimens, Eosinophilic intracy- If available, in situ hybridization may
but also skin biopsies (such toplasmic and intra- be most sensitive for detection of
as footpad biopsies) nuclear inclusions; virus in tissues.
CDV antigen with
immunostaining or
CDV RNA with in
situ hybridization

CDV, Canine distemper virus; RT, reverse transcriptase; SN, serum neutralization.
160 SECTION 1  Viral Diseases

and lymphoplasmacytic perivascular cuffing may be present. The


virus has a predilection for white matter of the lateral cerebellar
peduncles, the cerebellum, and the dorsolateral medulla adjacent
to the fourth ventricle, but lesions can also occur elsewhere, such
as the thalamus, midbrain, and pyriform lobes of the cerebral
cortex. Less commonly, nonsuppurative meningitis is observed.
Infection of the lungs leads to a lymphocytic and histiocytic inter-
stitial pneumonia with proliferation of the alveolar epithelium;
neutrophilic bronchopneumonia occurs as a result of secondary
bacterial infection. There is often widespread lymphoid deple-
tion and necrosis in all reticuloendothelial tissues (see Figure
15-3), although lymphoid hyperplasia may be observed in dogs
with chronic distemper. Epithelial cell necrosis may be noted in
the dental ameloblast layer, trachea, and bladder mucosa.
The diagnosis of CDV infection is supported by the identifi-
cation of eosinophilic intracytoplasmic or intranuclear inclusion
bodies, which are 1 to 5 µm in diameter and occur in a variety of
cell types, but especially in neurons, astrocytes, microglial cells,
conjunctival epithelial cells, bladder epithelium, footpad epi-
thelium, and sometimes lymphocytes (Figure 15-8). Less com-
monly, syncytia may be identified, such as within the meninges,
and lymph nodes. Confirmation of the presence of CDV in tis-
FIGURE 15-8  Bronchial epithelium from a dog infected with CDV. Intracytoplas- sues can be established with the use of immunostaining, in situ
mic inclusions are abundant (arrows). H&E stain. (Image courtesy Dr. Patricia Pesavento, hybridization, or RT-PCR (Figure 15-9).
University of California, Davis Veterinary Anatomic Pathology service.)
Treatment and Prognosis
When required, the mainstay of treatment for CDV infection is
supportive care. No specific antiviral drug is available for treat-
ment of CDV infection, although ribavirin and 5-ethynyl-1-β-
d-ribofuranosylimidazole-4-carboxamide (EICAR) have shown
efficacy in  vitro43 and for treatment of measles in a rodent
model.44 The pharmacokinetics and toxicity of these drugs in
dogs require further study.
Dogs with mild respiratory or gastrointestinal distemper
may recover spontaneously without treatment. Dogs with
severe respiratory and gastrointestinal disease may require hos-
pitalization and treatment with IV fluids, antimicrobial drugs
for secondary bacterial pneumonia, oxygen supplementation,
nebulization, and coupage. Ideally, these dogs should be housed
in isolation, although in many veterinary hospitals, isolation
may not permit treatment with oxygen and adequate monitor-
ing. Under these circumstances, dogs could be housed under
strict barrier precautions where oxygen and critical care can be
provided (see Chapter 11). When possible, collection of a trans-
tracheal lavage specimen for culture and susceptibility is recom-
FIGURE 15-9  Immunohistochemistry that shows CDV antigen (brown staining) in mended for dogs with secondary bacterial pneumonia. When
bladder transitional epithelial cells from a 2-year-old male neutered Australian cattle dog this is not possible, the use of antimicrobial drugs likely to be
infected with CDV that had been recently adopted from a shelter and developed cough, effective against Bordetella bronchiseptica (such as doxycycline)
ataxia, blindness, and seizures. is recommended (see Chapter 38). Dogs with severe broncho-
pneumonia may require treatment with parenteral broad-spec-
hemorrhages and meningeal congestion. In some dogs, gross trum antimicrobial drug combinations, such as ampicillin and
necropsy abnormalities are minimal. Concurrent external and a fluoroquinolone. Other supportive treatments include enteral
intestinal parasitic infections may also be identified. nutrition, artificial tears for dogs with KCS, and antiemetics.
Parenteral nutrition should be used only if absolutely necessary,
Histopathologic Findings because of the immunosuppression that occurs with CDV infec-
Histopathologic findings in the brain and spinal cord are vari- tion and possibility of nosocomial infections. Vitamin A sup-
able and depend on the CDV strain, the immune status of the plementation before experimental infection reduced morbidity
host, the presence of secondary infections, and disease chro- and mortality associated with CDV infection in ferrets,45 but its
nicity. Findings in the brain and spinal cord include neuronal usefulness as a treatment for canine distemper requires further
necrosis and degeneration, and demyelination with subacute to investigation. Administration of daily oral vitamin A for 2 days
chronic infection. Multifocal gliosis, astrocytosis, vacuolization, to children younger than 2 years of age with measles reduces the
CHAPTER 15  Canine Distemper Virus Infection 161

risk of mortality and pneumonia-specific mortality.46 Ascorbic that vaccine virus shed by the dam may cause disease in the
acid has also been advocated as a treatment for distemper in pups.49 Postvaccinal encephalitis usually occurs 3 to 20 days
dogs, but its efficacy remains unproven.41 after vaccination and has been associated with certain batches
Owners of dogs with respiratory and gastrointestinal illness of vaccines that contain the Rockborn strain.50 Neurologic signs
should be warned that CNS signs have the potential to develop such as ataxia may be reversible in some dogs. Seizures may be
weeks to even months after apparent recovery. Unfortunately, progressive and irreversible. Some distemper vaccines available
the development of CNS signs is unpredictable. When neuro- on the market still contain the Rockborn strain, and Rockborn-
logic signs occur, they rarely resolve and may be progressive. The like viruses have been isolated from dogs with distemper, which
prognosis for dogs with severe neurologic signs is poor, but some may represent residual virulence of vaccine virus or circulation
neurologic signs, such as mild myoclonus, may still be compatible of vaccine-derived Rockborn-like viruses in the dog popula-
with normal daily activities and do not interfere with quality of tion.51 Attenuated live CDV vaccines have been incriminated
life. Euthanasia is recommended for dogs with neurologic signs as a cause of hypertrophic osteodystrophy, especially in young
that are progressive and not compatible with a good quality of life. Weimaraner dogs,41,52 but there is a paucity of published evi-
A single IV dose of dexamethasone or tapering antiinflam- dence that supports the association (see Chapter 12).
matory doses of glucocorticoids has been advocated to halt or A canarypox-vectored recombinant vaccine that expresses
reduce the severity of progressive CNS signs,41 although con- the H and F glycoproteins of CDV is also available for preven-
trolled prospective studies are required to fully evaluate the tion of canine distemper (Merial Limited). This vaccine induces
usefulness of this treatment in dogs with distemper. Anticon- a 3-year duration of immunity, protects dogs from severe disease
vulsants such as diazepam and, for chronic management, potas- when administered hours before shelter admission, can protect
sium bromide or phenobarbital can be used to treat seizures and puppies from virulent CDV infection in the face of maternal
prevent the spread of a seizure focus. However, they do not pre- antibody, and can be used in pregnant bitches and puppies as
vent progressive CDV infection of the CNS and are sometimes young as 4 weeks of age in the face of an outbreak or in heavily
poorly effective. Mexiletine and procainamide have also been contaminated environments.48,53 The recombinant vaccine has
suggested as treatments for distemper myoclonus, but their true replaced the use of canine measles vaccines, which were previ-
efficacy is unreported. To date, the author has had disappoint- ously used to overcome maternal antibody in the face of dis-
ing results using these treatments in a limited number of dogs temper outbreaks. Whether the recombinant vaccine overcomes
with persistent distemper myoclonus. maternal antibody that is directed against the canarypox vector
vaccine virus as effectively as it overcomes maternal antibody
Immunity and Vaccination against the attenuated CDV vaccine virus requires further evalu-
ation. Further study of the relative efficacy of the recombinant
Immunity to CDV requires antibodies as well as cell-mediated and attenuated live vaccines in heavily contaminated environ-
immunity. Recovery from natural infection provides lifelong ments such as shelters is also required. The canarypox vectored
immunity. Distemper can be effectively prevented through recombinant vaccine is also the vaccine of choice for ferrets,
immunization with attenuated live or recombinant vaccines. and a specific ferret vaccine is available at the time of writing
Inactivated vaccines do not provide sufficient protection and (Purevax Ferret Distemper, Merial Limited).
are not available in the United States. In contrast, proper vac- Vaccine strains of CDV, such as Onderstepoort, Snyder
cination with attenuated live vaccines can provide long-lasting Hill, and Lederle, belong to the America-1 lineage, but field
(at least 3 to 5 years), sterile immunity.47 Attenuated live vac- strains that circulate in North America belong primarily to
cines should be given every 3 to 4 weeks, commencing no earlier the ­America-2 lineage. Concern has been raised that these vac-
than 6 weeks of age. Maternal antibodies are usually absent cines may not adequately protect against infection with field
by 12 to 14 weeks of age, and so the last vaccine should be strains, but cross-neutralization studies suggest that antigenic
given no sooner than 14 to 16 weeks of age (consider exten- differences are not sufficient to warrant changes in the cur-
sion to 16 to 20 weeks in breeding kennels and shelters).47 Ide- rent vaccines. The development of distemper in vaccinated
ally pups should be isolated for at least 7 to 10 days after the dogs may reflect failure of vaccine efficacy as a result of the
last vaccine is administered (see Appendix). For dogs older than improper administration of vaccines, improperly timed vacci-
16 weeks of age that are brought to the veterinarian for their nation series, or the use of vaccines that have been stored and
first vaccination, two doses of vaccine should be given 3 to handled inappropriately. The last is especially important for
4 weeks apart, although a single dose may provide protection. CDV vaccines because attenuated live vaccine virus is so labile.
A booster is recommended at 1 year and then no earlier than Vaccines should be stored in a refrigerator that maintains an
every 3 years thereafter.47 For shelter dogs, vaccination should internal temperature of 0°C to 4°C and reconstituted with the
be performed as soon as possible before entry into the shelter. proper diluent immediately (within 1 hour) before administra-
A single vaccine given even hours before entry can provide pro- tion. It has been recommended that vaccine held for longer than
tection from severe disease.48 Infection can still occur, but dis- 1 hour after reconstitution be discarded.47 A second dose should
ease severity is lessened. be given if some of the vaccine is found on the haircoat after
Although generally safe and effective when used according to administration.
label recommendations, attenuated live CDV vaccine virus can
cause encephalitis when administered to immunocompromised Prevention
dogs and puppies less than 6 weeks of age, and severe disease
can occur in susceptible species such as ferrets and certain wild- Properly timed vaccination is the key to prevention of CDV
life species. Administration of attenuated live CDV vaccines to infection. Puppies with declining maternal antibody titers should
dams immediately after whelping has been associated with the be kept away from other dogs until the vaccination series is
development of encephalitis in the puppies, and it was suggested complete. In shelter situations, dogs with signs of respiratory or
162 SECTION 1  Viral Diseases

gastrointestinal disease should be held well away from other dogs Public Health Aspects
(at least 25 feet has been suggested37) and handled only after
apparently healthy dogs have been cared for. Puppies should In general, CDV is not believed to infect human cells or cause
always be separated from adult dogs using equipment desig- disease in people. However, both CDV and measles virus have
nated only for that area, and overcrowding should be prevented. played a controversial role in Paget’s disease of bone, a com-
Quarantine for CDV is difficult because the incubation period mon skeletal disorder characterized by focal abnormalities of
for development of neurologic signs may be as long as 6 weeks. increased bone turnover, which is associated with bone pain,
Wildlife species such as raccoons can also shed CDV and were deformity, and pathologic fractures.55 Paget’s disease most often
suspected as a source of infection in an outbreak that occurred in occurs in the elderly, men are more likely to be affected than
a Chicago shelter, when raccoons and dogs were co-transported women, and the overall prevalence of the disease has decreased
in the back of a vehicle.37 Routine cleaning and disinfection of in many geographic locations.56 Genetic factors clearly play a
contaminated surfaces readily eliminates virus in the environ- role in the disease, but in some studies, the RNA of paramyxovi-
ment. Proper nutrition may also help to prevent disease due to ruses that include CDV has been detected within lesions.57 Other
CDV. Isolated vitamin A deficiency is a major risk factor for studies have failed to detect the nucleic acid of CDV in lesions,
severe measles and death in humans and distemper in ferrets. Ret- and contamination has been suggested as a reason for positive
inoids interfere with replication of measles virus in vitro through test results.55 However, CDV has been shown to infect and rep-
a type I IFN-dependent mechanism.54 The impact of nutritional licate in human osteoclast precursors, further raising concern
status on disease due to CDV requires further investigation. about the possibility of zoonotic transmission of CDV.58

CASE EXAMPLE Cardiovascular, respiratory, gastrointestinal, and


urogenital systems: No clinically significant findings were
identified. All peripheral lymph nodes were normal sized.
Signalment: “Molly”, a 1.5-year-old female spayed golden Average systolic blood pressure (10 measurements) was 120
retriever from Sacramento, CA mm Hg.
History: Molly was evaluated for a 1-month-long history Neurologic Examination:
of abnormal behavior that included stumbling, failure Mentation: Obtunded and not responsive to verbal and most
to catch balls, and loss of interest in activity. She also had tactile stimuli; head pressing.
a 1-year history of seizures, which were thought to be Gait/Posture: Compulsive circling and leaning to the left was
due to idiopathic epilepsy and which were treated with present.
phenobarbital (4 mg/kg PO q12h) and potassium bromide Cranial nerves: No clinically significant abnormalities were
(17 mg/kg PO q12h). This was associated with a decrease in identified.
seizure frequency. Two weeks after the onset of abnormal Postural reactions: Placing reactions were absent in the left
behavior, Molly was taken to a local veterinary clinic. A thoracic and left and right pelvic limbs.
serum phenobarbital level at that time was 50 µg/mL Segmental reflexes: Diminished biceps and withdrawal
(therapeutic range, 10 to 40 µg/mL). The phenobarbital reflexes were present, left thoracic limb only.
dose was decreased (1.4 mg/kg PO q24h), but Molly began Panniculus: No abnormalities detected.
circling to the left, head pressing, pacing, and vocalizing. She Spinal palpation: Pain was elicited in several regions along
also stopped responding to the owners when they called the spine. Resisted cervical ventroflexion.
her name, and two episodes of vomiting were noted. In the Neuroanatomic localization: Diffuse cerebral or thalamic
week before evaluation, Molly refused to eat dry food, but disease; C6 to T2 myelopathy.
ate canned dog food. Her thirst had decreased and she was Laboratory Findings:
voluntarily urinating in the house. A serum phenobarbital CBC:
level was repeated 2 days before she was evaluated and was HCT 37.9% (40-55%)
13 µg/mL. Molly had been immunized regularly (CDV, CPV, MCV 68 fL (65-75 fL)
CAV-2, and parainfluenza) and received monthly flea, tick, MCHC 35.4 g/dL (33-36 g/dL)
and heartworm prophylaxis. WBC 7510 cells/µL (6000-13,000 cells/µL)
Current medications: Phenobarbital (1.4 mg/kg PO q24h); Neutrophils 4859 cells/µL (3000-10,500 cells/µL)
potassium bromide (17 mg/kg PO q12h). Lymphocytes 2155 cells/µL (1000-4000 cells/µL)
Physical Examination: Monocytes 338 cells/µL (150-1200 cells/µL)
Body weight: 23.2 kg 427,000 platelets/µL (150,000-400,000 platelets/µL).
General: Obtunded but hydrated. T = 101.7°F (38.7°C), HR = 60 Serum Chemistry Profile:
beats/min, RR = 36 breaths/min, mucous membranes pink, Sodium 141 mmol/L (145-154 mmol/L)
CRT = 1 sec. Potassium 4.4 mmol/L (4.1-5.3 mmol/L)
Integument, eyes, ears, nose and throat: No clinically Chloride 109 mmol/L (105-116 mmol/L)
significant abnormalities were identified. A full Bicarbonate 21 mmol/L (16-26 mmol/L)
ophthalmologic examination was also unremarkable. Phosphorus 3.9 mg/dL (3.0-6.2 mg/dL)
Musculoskeletal: Body condition score was 5/9. Molly was Calcium 12.2 mg/dL (9.9-11.4 mg/dl)
ambulatory on all four limbs. BUN 6 mg/dL (8-31 mg/dL)
CHAPTER 15  Canine Distemper Virus Infection 163

Creatinine 0.6 mg/dL (0.5-1.6 mg/dL) Lumbar Cisternal


Glucose 109 mg/dL (70-118 mg/dL)
Total protein 6.1 g/dL (5.4-7.4 g/dL) Large mononu- 8 20
Albumin 2.7 g/dL (2.9-4.2 g/dL) clear cells (%)
Globulin 3.4 g/dL (2.3-4.4 g/dL) Microscopic Mild to ­moderate Marginal
ALT 132 U/L (19-67 U/L) evaluation lymphocytic mononuclear
AST 62 U/L (21-54 U/L) reactivity (lymphocytic)
ALP 155 U/L (15-127 U/L) pleocytosis
Creatine kinase 399 U/L (46-320 U/L)
Gamma GT 10 U/L (0-6 U/L) Treatment and Outcome: Molly was hospitalized for a
Cholesterol 291 mg/dL (135-345 mg/dL) week during the diagnostic work-up and treated with
Total bilirubin 0.2 mg/dL (0-0.4 mg/dL). intravenous fluids and potassium bromide (35 mg/kg PO
Serum ionized calcium: 1.39 mmol/L q12h). She became progressively more obtunded and
Urinalysis: SGr 1.014; pH 7.0, few amorphous crystals. No other showed additional neurologic signs of circling to the right
abnormalities detected. and positional horizontal nystagmus with a fast phase
Serum bromide concentration: 1.1 mg/mL (therapeutic to the right. The narrowed disc spaces were thought to
range, 1.0 to 2.0 mg/mL) represent intervertebral disc disease, and granulomatous
Serum phenobarbital concentration: 1.4 µg/mL (therapeutic meningoencephalitis was suspected. Potassium bromide
range, 15 to 40 µg/mL) was discontinued, and treatment with phenobarbital (5 mg/
Serum bile acids: Pre-meal, 14 µmol/L (reference range, kg PO q12h) reinstituted. Dexamethasone was administered
0-12 µmol/L); post-meal, 16 µmol/L (reference range, 0-16 (0.2 mg/kg, IV q24h), but there was minimal improvement,
µmol/L) and euthanasia was elected.
Imaging Findings: Necropsy Findings:
Thoracic radiographs: No clinically significant abnormalities • Brain, cerebrum: severe, generalized, chronic neuronal loss,
were present. axonal necrosis, and myelin degeneration with abundant
Spinal radiographs: Disc space narrowing was present eosinophilic inclusion bodies and canine distemper antigen
between the sixth and seventh cervical vertebrae and to (immunohistochemistry)
a greater extent between the seventh cervical and first • Brain and spinal cord (C1 to T7), corticospinal tracts: severe,
thoracic vertebrae. diffuse axonal necrosis and myelin degeneration
Abdominal ultrasound: No significant lesions were identified. • Brain, cerebrum, and meninges: generalized, lymphoplas-
Magnetic resonance imaging: Precontrast: Multiple small macytic, perivascular cuffing
regions of hyperintensity were noted on dual echo and • Tonsil, lymph nodes (mandibular, mesenteric, thoracic):
FLAIR sequences, one of which was in the left thalamus mild, multifocal lympholysis with intranuclear and intracy-
along the third ventricle and another diffusely along the toplasmic eosinophilic inclusion bodies and canine distem-
lateral aspect of the right thalamus. There was a linear region per antigen
of hyperintensity along the right parietal and temporal Diagnosis: Chronic distemper encephalitis with demyelination
lobes. On T1-weighted postcontrast images, a small focus (“old dog encephalitis”)
of contrast enhancement was noted in the ventral aspect Comments: Distemper was not suspected in this dog
of the right thalamus in the region of hyperintensity on because of the dog’s older age and history of vaccination.
the precontrast fluid-weighted sequences. Impressions: Initial infection may have occurred before vaccination
Multifocal, relatively non–contrast-enhancing lesions in was complete, vaccination may have been improperly
the cerebrum and thalamus suggestive of cerebral edema, performed, or the dog may have been a vaccine
encephalitis, or small infarcts. nonresponder, which is believed to occur rarely. The
Electroencephalography: No abnormalities were detected. previous diagnosis of epilepsy further complicated the
CSF Analysis: identification of distemper as an underlying cause. This case
Lumbar Cisternal example highlights the importance of including distemper
on the differential diagnosis list for young adult dogs with
Protein (mg/dL) 31 21
neurologic signs, because of the chronic course of disease
Total RBC (cells/ 111 2 that can occasionally occur. Additional laboratory tests
µL) that might or might not have been helpful for diagnosis
Total nucleated 1 3 of distemper in this dog include comparison of serum
cells (cells/µL) antibody titers to CDV in the CSF and serum, and RT-PCR for
CDV on CSF. Neurologic signs were progressive in this dog
Neutrophils (%) 0 0
and largely unresponsive to anticonvulsant drugs, and so
Small mononu- 92 80 the prognosis was extremely poor.
clear cells (%)
164 SECTION 1  Viral Diseases

SUGGESTED READINGS 19. Bittegeko SB, Arnbjerg J, Nkya R, et al. Multiple dental develop-
mental abnormalities following canine distemper infection. J Am
Beineke A, Puff C, Seehusen F, et al. Pathogenesis and immunopathol- Anim Hosp Assoc. 1995;31:42-45.
ogy of systemic and nervous canine distemper. Vet Immunol Immuno- 20. Boutoille FF, Hennet PR. Diagnostic imaging in veterinary dental prac-
pathol. 2009;127:1-18. tice. Enamel hypoplasia. J Am Vet Med Assoc. 2011;238:1251-1253.
Newbury S, Larson LJ, Schultz RD. Canine distemper virus. In: 21. Schobesberger M, Summerfield A, Doherr MG, et al. Canine dis-
Miller L, Hurley K, eds. Infectious disease management in animal shel- temper virus–induced depletion of uninfected lymphocytes is associated
ters. Ames, IA: Wiley-Blackwell; 2009:161-172. with apoptosis. Vet Immunol Immunopathol. 2005;104:33-44.
22. Kerdiles YM, Cherif B, Marie JC, et  al. Immunomodulatory
REFERENCES properties of morbillivirus nucleoproteins. Viral Immunol.
1. Gamiz C, Martella V, Ulloa R, et al. Identification of a new geno- 2006;19:324-334.
type of canine distemper virus circulating in America. Vet Res 23. Ehrensperger F, Pospischil A. Spontaneous mixed infections with dis-
Commun. 2011;35:381-390. temper virus and Toxoplasma in dogs. Dtsch Tierarztl Wochenschr.
2. Panzera Y, Gallo Calderón M, Sarute N, et  al. Evidence of two 1989;96:184-186.
co-circulating genetic lineages of canine distemper virus in South 24. Smith HW, Buxton A. Incidence of salmonellae in faces of dogs
America. Virus Res. 2012;163:401-404. suffering from distemper. Nature. 1950;166:824.
3. Kapil S, Allison RW, Johnston 3rd L, et al. Canine distemper virus 25. Fawi MT. Tag el Din MH, el-Sanousi SM. Canine distemper as a
strains circulating among North American dogs. Clin Vaccine predisposing factor for Nocardia asteroides infection in the dog. Vet
Immunol. 2008;15:707-712. Rec. 1971;88:326-328.
4. Amude AM, Alfieri AA, Alfieri AF. Clinicopathological findings in 26. Ribeiro MG, Salerno T, Mattos-Guaraldi AL, et al. Nocardiosis: an
dogs with distemper encephalomyelitis presented without charac- overview and additional report of 28 cases in cattle and dogs. Rev
teristic signs of the disease. Res Vet Sci. 2007;82:416-422. Inst Med Trop Sao Paulo. 2008;50:177-185.
5. Blixenkrone-Moller M, Svansson V, Have P, et al. Studies on mani- 27. Dyer NW, Schamber GJ. Pneumocystosis associated with
festations of canine distemper virus infection in an urban dog popu- canine distemper virus infection in a mink. Can Vet J. 1999;40:
lation. Vet Microbiol. 1993;37:163-173. 577-578.
6. Richards TR, Whelan NC, Pinard CL, et al. Optic neuritis caused 28. Lemberger KY, Gondim LF, Pessier AP, et al. Neospora caninum
by canine distemper virus in a Jack Russell terrier. Can Vet J. infection in a free-ranging raccoon (Procyon lotor) with concurrent
2011;52:398-402. canine distemper virus infection. J Parasitol. 2005;91:960-961.
7. Yanagi Y, Takeda M, Ohno S. Measles virus: cellular receptors, 29. Baumgartner W, Boyce RW, Alldinger S, et al. Metaphyseal bone
tropism and pathogenesis. J Gen Virol. 2006;87:2767-2779. lesions in young dogs with systemic canine distemper virus infec-
8. Beineke A, Puff C, Seehusen F, et al. Pathogenesis and immunopa- tion. Vet Microbiol. 1995;44:201-209.
thology of systemic and nervous canine distemper. Vet Immunol 30. Mee AP, Gordon MT, May C, et al. Canine distemper virus tran-
Immunopathol. 2009;127:1-18. scripts detected in the bone cells of dogs with metaphyseal osteopa-
9. von Messling V, Milosevic D, Cattaneo R. Tropism illumi- thy. Bone. 1993;14:59-67.
nated: lymphocyte-based pathways blazed by lethal morbillivi- 31. Bathen-Noethen A, Stein VM, Puff C, et  al. Magnetic resonance
rus through the host immune system. Proc Natl Acad Sci U S A. imaging findings in acute canine distemper virus infection. J Small
2004;101:14216-14221. Anim Pract. 2008;49:460-467.
10. von Messling V, Svitek N, Cattaneo R. Receptor (SLAM [CD150]) 32. Soma T, Ishii H, Hara M, et  al. Detection of canine distemper
recognition and the V protein sustain swift lymphocyte-based inva- virus antigen in canine serum and its application to diagnosis. Vet Rec.
sion of mucosal tissue and lymphatic organs by a morbillivirus. 2003;153:499-501.
J Virol. 2006;80:6084-6092. 33. An DJ, Kim TY, Song DS, et al. An immunochromatography assay
11. Pratakpiriya W, Seki F, Otsuki N, et  al. Nectin4 is an epithelial for rapid antemortem diagnosis of dogs suspected to have canine
cell receptor for canine distemper virus involved in neurovirulence. distemper. J Virol Methods. 2008;147:244-249.
J Virol. 2012;86:10207-10210. 34. Kim D, Jeoung SY, Ahn SJ, et al. Comparison of tissue and fluid
12. Appel MJ. Pathogenesis of canine distemper. Am J Vet Res. samples for the early detection of canine distemper virus in experi-
1969;30:1167-1182. mentally infected dogs. J Vet Med Sci. 2006;68:877-879.
13. Pearce-Kelling S, Mitchell WJ, Summers BA, et  al. Virulent and 35. Elia G, Decaro N, Martella V, et  al. Detection of canine dis-
attenuated canine distemper virus infects multiple dog brain cell temper virus in dogs by real-time RT-PCR. J Virol Methods.
types in vitro. Glia. 1991;4:408-416. 2006;136:171-176.
14. Rudd PA, Bastien-Hamel LE, von Messling V. Acute canine dis- 36. Si W, Zhou S, Wang Z, et al. A multiplex reverse transcription-nested
temper encephalitis is associated with rapid neuronal loss and local polymerase chain reaction for detection and differentiation of wild-
immune activation. J Gen Virol. 2010;91:980-989. type and vaccine strains of canine distemper virus. Virol J. 2010;7:86.
15. Summers BA, Appel MJ. Aspects of canine distemper virus and 37. Newbury S, Larson LJ, Schultz RD. Canine distemper virus. In:
measles virus encephalomyelitis. Neuropathol Appl Neurobiol. Miller L, Hurley K, eds. Infectious disease management in animal shel-
1994;20:525-534. ters. Ames, IA: Wiley-Blackwell; 2009:161-172.
16. Headley SA, Amude AM, Alfieri AF, et  al. Molecular detection 38. von Messling V, Harder TC, Moennig V, et al. Rapid and sensitive
of canine distemper virus and the immunohistochemical charac- detection of immunoglobulin M (IgM) and IgG antibodies against
terization of the neurologic lesions in naturally occurring old dog canine distemper virus by a new recombinant nucleocapsid pro-
encephalitis. J Vet Diagn Invest. 2009;21:588-597. tein-based enzyme-linked immunosorbent assay. J Clin Microbiol.
17. Wyss-Fluehmann G, Zurbriggen A, Vandevelde M, et al. Canine 1999;37:1049-1056.
distemper virus persistence in demyelinating encephalitis by 39. Noon KF, Rogul M, Binn LN, et al. Enzyme-linked immunosorbent
swift intracellular cell-to-cell spread in astrocytes is controlled assay for evaluation of antibody to canine distemper virus. Am J
by the viral attachment protein. Acta Neuropathol. 2010;119: Vet Res. 1980;41:605-609.
617-630. 40. Johnson GC, Fenner WR, Krakowka S. Production of immuno-
18. de Almeida DE, Roveratti C, Brito FL, et al. Conjunctival effects globulin G and increased antiviral antibody in cerebrospinal fluid
of canine distemper virus–induced keratoconjunctivitis sicca. Vet of dogs with delayed-onset canine distemper viral encephalitis.
Ophthalmol. 2009;12:211-215. J Neuroimmunol. 1988;17:237-251.
CHAPTER 15  Canine Distemper Virus Infection 165

41. Greene CE, Appel MJ. Canine distemper. In: Greene CE, ed. Infec- 50. Cornwell HJ, Thompson H, McCandlish IA, et al. Encephalitis in
tious diseases of the dog and cat. 3rd ed. St. Louis, MO: Saunders dogs associated with a batch of canine distemper (Rockborn) vac-
Elsevier; 2006:25-41. cine. Vet Rec. 1988;122:54-59.
42. Litster AL, Pressler B, Volpe A, et al. Accuracy of a point-of-care 51. Martella V, Blixenkrone-Moller M, Elia G, et al. Lights and shades
ELISA test kit for predicting the presence of protective canine par- on an historical vaccine canine distemper virus, the Rockborn
vovirus and canine distemper virus antibody concentrations in strain. Vaccine. 2011;29:1222-1227.
dogs. Vet J. 2012;193:363-366. 52. Harrus S, Waner T, Aizenberg, et al. Development of hypertrophic
43. Dal Pozzo F, Galligioni V, Vaccari F, et  al. Antiviral efficacy of osteodystrophy and antibody response in a litter of vaccinated Wei-
EICAR against canine distemper virus (CDV) in vitro. Res Vet Sci. maraner puppies. J Small Anim Pract. 2002;43:27-31.
2010;88:339-344. 53. Larson LJ, Schultz RD. Three-year duration of immunity in dogs
44. Wyde PR, Moore-Poveda DK, De Clercq E, et  al. Use of cotton vaccinated with a canarypox-vectored recombinant canine distem-
rats to evaluate the efficacy of antivirals in treatment of mea- per virus vaccine. Vet Ther. 2007;8:101-106.
sles virus infections. Antimicrob Agents Chemother. 2000;44: 54. Trottier C, Colombo M, Mann KK, et  al. Retinoids inhibit mea-
1146-1152. sles virus through a type I IFN-dependent bystander effect. FASEB J.
45. Rodeheffer C, von Messling V, Milot S, et al. Disease manifesta- 2009;23:3203-3212.
tions of canine distemper virus infection in ferrets are modulated 55. Ralston SH, Afzal MA, Helfrich MH, et  al. Multicenter blinded
by vitamin A status. J Nutr. 2007;137:1916-1922. analysis of RT-PCR detection methods for paramyxoviruses in rela-
46. Huiming Y, Chaomin W, Meng M. Vitamin A for treating measles tion to Paget’s disease of bone. J Bone Miner Res. 2007;22:569-577.
in children. Cochrane Database Syst Rev. 2005:CD001479. 56. Cooper C, Harvey NC, Dennison EM, et al. Update on the epide-
47. Welborn LV, DeVries JG, Ford R, et al. 2011 AAHA canine vac- miology of Paget’s disease of bone. J Bone Miner Res. 2006;21(suppl
cination guidelines. J Am Anim Hosp Assoc. 2011;47:1-42. 2):P3-P8.
48. Larson LJ, Schultz RD. Effect of vaccination with recombinant 57. Mee AP, Dixon JA, Hoyland JA, et  al. Detection of canine dis-
canine distemper virus vaccine immediately before exposure under temper virus in 100% of Paget’s disease samples by in situ-reverse
shelter-like conditions. Vet Ther. 2006;7:113-118. transcriptase-polymerase chain reaction. Bone. 1998;23:171-175.
49. McCandlish IA, Cornwell HJ, Thompson H, et  al. Distem- 58. Selby PL, Davies M, Mee AP. Canine distemper virus induces
per encephalitis in pups after vaccination of the dam. Vet Rec. human osteoclastogenesis through NF-kappaB and sequestosome
1992;130:27-30. 1/P62 activation. J Bone Miner Res. 2006;21:1750-1756.
CHAPTER 16

Canine Herpesvirus Infection


Autumn P. Davidson

results either in late-term abortion of a litter or neonatal death


Overview of Canine Herpesvirus Infection within the first few weeks of life, because inadequate peripartu-
First Described: New York State, United States, 19651 rient maternal antibodies exist to allow passive immunity to be
Cause: Canine herpesvirus-1 (Family Herpesviridae) acquired by the neonates. Puppies born to a naïve bitch may also
become infected as a result of contact with other dogs that shed
Affected Hosts: Domestic and wild Canidae the organism. The incubation period of CHV-1 disease is 6 to
Geographic Distribution: Worldwide 10 days. The virus replicates in the epithelial cells of the orona-
sal and pharyngeal mucosa, the genital tract, and the regional
Mode of Transmission: Direct oronasal contact with infected
lymphatics. Replication and spread of the virus is facilitated
secretions, transplacental transmission
by the presence of a low body temperature (<38°C or 100°F),
Major Clinical Signs: Abortion in bitches; upper respiratory which is normal for canine neonates.
disease and keratitis in adult dogs; in neonates, inappe- Older (>3 to 5 weeks of age) puppies that are exposed to
tence, failure to thrive, vocalization, tachypnea, diarrhea, CHV-1 may develop subclinical infection, or the course of dis-
neurologic signs, abdominal pain, nasal discharge, weight ease is less severe, as a result of their ability to mount a febrile
loss, mucosal petechiation, death response. Latent infection also may develop. Concerns have
Differential Diagnoses: Bacterial sepsis, infections with been raised about latency and the possibility of late develop-
other viruses such as canine distemper virus and canine ment of neurologic signs.3
parvovirus Clinical signs are more likely to occur in animals that are
hypothermic or immunosuppressed. Signs in the neonate are not
Human Health Significance: Canine herpesvirus-1 does not
specific and include incessant vocalization, anorexia (with poor
infect humans.
weight gain), dyspnea, abdominal pain, incoordination, diar-
rhea, serous to hemorrhagic nasal discharge, and petechiation
of the mucous membranes. The mortality rate in litters infected
Etiology and Epidemiology in utero or during birth can approach 100%, with deaths occur-
ring during the first few days to a week of life. Exposed, surviv-
Canine herpesvirus (CHV-1) is an enveloped virus that belongs ing older neonates may develop a late onset of central nervous
to the family Herpesviridae (Figure 16-1). It has been reported signs including blindness, ataxia, and deafness; apparent com-
from the United States, Canada, Australia, Japan, England, and plete recovery has also been reported.
Germany. CHV-1 was first recognized in the mid-1960s in asso- The recently infected brood bitch generally shows no clini-
ciation with a fatal disease in puppies.2 The virus is commonly cal signs. Healthy adult dogs of either gender can develop mild
blamed for acute neonatal puppy death or failure to thrive, upper respiratory signs (sneezing, serous oculonasal discharge,
sometimes termed the “fading puppy syndrome.” When con- keratitis) for a few days but are otherwise usually clinically
firmed as a cause of disease, untreated CHV-1 infection in neo- unaffected.4-6 Additional information on respiratory and ocular
nates can cause high (up to 100%) mortality among littermates. disease caused by CHV-1 can be found in Chapter 17.
The virus is temperature sensitive and prefers to replicate at
temperatures less than 37°C. It is not stable in the environment Diagnosis
and is readily inactivated by disinfectants. As with other herpes-
viral infections, recovery from disease is associated with lifelong Antemortem diagnosis of CHV-1 infection in neonates can
latent infection of the neural ganglia, with periodic reactivation be challenging. Necropsy of a deceased littermate is usually
of shedding in association with stress or immunosuppression, required.
such as that which results from overcrowding and pregnancy.
CHV-1 infection has not been reported in cats. Virus Isolation and Molecular Diagnosis Using
the Polymerase Chain Reaction
Clinical Features Tissues obtained at necropsy can be submitted for virus isolation
or molecular diagnosis using the PCR. Diagnosis by virus isola-
Signs and Their Pathogenesis tion takes days. CHV-1–specific PCR assays can also confirm
Transmission of CHV-1 can occur subsequent to close contact infection, in a more timely fashion. Clinicians should confirm
with infectious vaginal fluids during whelping or with vulvar the availability and turnaround time of commercially available
or oronasal secretions in the postpartum period. Exposure of diagnostic tests when attempting to investigate a possible out-
a naïve bitch to CHV-1 during the last 3 weeks of gestation break of CHV-1 infection in puppies.7-9

166
CHAPTER 16  Canine Herpesvirus Infection 167

Pathologic Findings pink and red due to coagulation necrosis (Figure 16-2, B). Mul-
Gross Pathologic Findings tifocal random acute necrosis of other organs, including the liver,
Gross changes in the kidneys at necropsy of pups infected with pancreas, intestine and adrenal glands, may also be apparent.
CHV-1 include multifocal petechial to ecchymotic subcapsular Lesions may resemble those of bacterial sepsis.
hemorrhages (Figure 16-2). The pleural surfaces may be mottled
Histopathologic Findings
Histopathology can be used to confirm CHV-1 infection but may
be unrewarding in the clinical scenario unless very timely. Charac-
teristic viral intranuclear inclusion bodies can be difficult to find.
Histopathologic evaluation commonly reveals severe nephrosis
(proximal renal tubular epithelial necrosis and renal pelvic epithelial
apoptosis with swollen vesicular nuclei); diffuse severe hepatic con-
gestion and inflammation with vacuolar hepatopathy (neutrophilic,
histiocytic, and lymphocytic infiltration with apoptosis); acute
severe necrotizing bronchointerstitial pneumonia with pulmonary
edema and hemorrhage (terminal bronchiolar hemorrhage and
edema with fibrin accumulation, macrophage infiltration, necrosis,
and alveolar disruption); and thymic, lymph node, and splenic lym-
pholysis (severe diffuse cortical lympholysis with sinus congestion).

Treatment
Treatment of puppies with CHV-1 infection is unrewarding and
rarely effective, and residual cardiac and neurologic damage
can occur in recovered animals. Treatment with immune serum
FIGURE 16-1  Structure of a herpesvirus. Herpesviruses are large, enveloped DNA from seroconverted dams is typically ineffective in sick puppies,
viruses. The DNA is contained within an icosahedral capsid.
and warming also has little effect.

C D
FIGURE 16-2  A, Kidneys from 12-day-old mastiff puppy from a litter of 17 puppies, 14 of which died. CHV-1 was isolated from the puppy. Gross changes include subcapsular hemor-
rhagic foci typical of (but not pathognomonic for) herpetic nephritis. On cut section, white and red cortical streaks are evident; a hemorrhagic medulla is evident. B, Multifocal pink and red
pleural mottling in the puppy described in A. Serosanguineous pleural fluid was present. Histopathology identified multiple areas of pulmonary necrosis and bronchopneumonia. C, Small-
intestinal serosal petechiation in the same puppy. Histopathology identified enterocyte necrosis consistent with CHV-1 infection. D, Liver of the same puppy showing uniform firmness
and diffuse mottle with rounded edges. Histopathology identified scattered multiple foci of acute hepatic coagulation necrosis; eosinophilic intranuclear inclusions typical of herpes viral
inclusions were seen within viable hepatocytes. (Images courtesy JR Peauroi.)
168 SECTION 1  Viral Diseases

An incompletely developed immune system and inadequate Immunity, Vaccination, and Prevention
thermoregulation during the first days of life means neonates
are vulnerable to systemic infection (by both bacterial and viral Minimal transplacental transfer of immunity occurs in the dog.
pathogens). The umbilicus of neonates should be treated with Adequate ingestion of colostrum must occur promptly postpar-
tincture of iodine immediately after birth to reduce contami- tum for puppies to acquire passive immunity. The transmission
nation and prevent bacterial ascent into the peritoneal cavity of protective passive immunity (placental or colostral antibodies)
(omphalitis-peritonitis). Neonatal bacterial peritonitis with bac- between a bitch and her puppies depends on the prior existence
teremia can cause rapid neonatal death if not recognized and of adequate serum maternal antibodies. Therefore, breeding
treated promptly (see Figure 34-1). Factors that predispose pup- bitches with exposure to CHV-1 earlier in life have the best
pies to bacteremia and sepsis include endometritis in the bitch, opportunity to seroconvert and develop protective antibodies.
a prolonged delivery/dystocia, feeding of replacement formulas, This commonly occurs in kennels or at crowded canine events
the use of ampicillin, stress, low birth weight (<350 g), and chill- such as dog shows and trials. Documentation of positive CHV-1
ing with body temperatures less than 35.5°C or 95°F. Bacterial serology generally indicates the presence of adequate maternal
organisms most frequently associated with bacteremia are Esche- antibodies.
richia coli, streptococci, staphylococci, and Klebsiella spp. Com- Bitches who are naïve to CHV-1 during pregnancy must be
monly, a decrease in weight gain, failure to suckle, hematuria, strictly isolated from potential exposure during gestation and
persistent diarrhea, unusual vocalization, abdominal distention for at least 6 weeks postpartum to prevent transmission of the
and pain, and sloughing of the extremities indicate sepsis may virus to her fetuses or neonates. Subsequent litters of the infected
be present. These signs are similar to those encountered with pregnant or postpartum bitch are usually normal because of
CHV-1 infection. Prompt therapy with broad-spectrum, bacte- acquisition of maternal antibodies.
ricidal antibiotics; optimal nutrition via supported nursing, tube Vaccine development has been hampered by the poor immu-
feeding, or bottle feeding; maintenance of body temperature; and nogenicity characteristic of the herpesviral vaccines devel-
appropriate fluid replacement are indicated. The third-generation oped for other species, such as feline herpesvirus-1 vaccines.
cephalosporin ceftiofur sodium is an appropriate choice for treat- In Europe, a vaccine (Eurican Herpes 205) has been available
ment of neonatal sepsis, because it alters normal intestinal flora since 2003. Administration of two doses of the vaccine is rec-
minimally and is usually active against the causative organisms. ommended, first during estrus or early pregnancy and again 1
Failure of neonatal sepsis to respond to antibacterial drug treat- to 2 weeks before whelping. No suggestion for an evaluation
ment should prompt consideration of CHV-1 infection. Specific anti- of the bitch’s serologic status prior to vaccination is made by
viral therapy, if instituted in a timely manner, may reduce mortality the manufacturer. Revaccination with two doses of the vaccine
from CHV-1. Acyclovir has activity against herpes simplex virus is recommended at each pregnancy. No independent, nonpro-
(see Chapter 7). Its use in veterinary medicine is not well established, prietary studies exist corroborating this vaccine’s efficacy or
and it should be used with caution and only in situations where benefit. The vaccine is not currently available in the United
indicated. The safety and effectiveness in humans less than 2 weeks States.12,13
of age is not established. The dose (20 mg/kg PO q6h for 5 days)
is extrapolated from that for humans.10 The efficacy of famciclovir Public Health Aspects
for treatment of CHV-1 infection requires study, but the pharmaco-
kinetics of famciclovir in dogs resembles that in humans,11 so much In general, herpesviruses are very host specific, and CHV-1 is
lower doses may be required when compared with cats. not known to infect humans.

CASE EXAMPLE ceftiofur sodium [2.5 mg/kg SC q12h]), the puppies died within
12 hours and then underwent necropsy examination. Tissues
collected for histopathologic evaluation included kidney, liver,
Two, 25-day-old, female Labrador retriever puppies were exam- lung, thymus, mesenteric lymph node, and spleen.
ined because of acute tachypnea and vocalization. The puppies Because of gross findings of hemorrhagic foci in the lungs
belonged to a litter of eight. Whelping occurred without inci- and kidneys, renal tissue was submitted for a CHV-1–specific
dent, and the pups showed normal weight gain and behavior PCR assay. The PCR assay later confirmed the presence of CHV-1
until the last 24 hours. Four days previously, the litter was vac- DNA in the renal tissue. Gross (foci of hemorrhage in the lungs
cinated with an intranasal Bordetella bronchiseptica vaccine and and kidneys) and histopathologic evaluations of the deceased
dewormed with pyrantel pamoate according to routine preven- puppies were typical of CHV-1 but were not confirmatory
tative health care practice. The litter nursed from birth, and the because of the similarity of findings to those of neonatal bac-
dam was normal. terial sepsis. Intranuclear inclusion bodies were suspected
Physical examination of both puppies revealed hypo- in macrophages from the lung of one puppy. Aerobic bacte-
thermia (rectal temperature 36.3°C [97°F] and 36.0°C [97°F], rial culture of the peritoneal cavity was negative for bacterial
respectively), tachypnea, a markedly tense abdomen, and growth.
continuous vocalization. Despite supportive care (exogenous Poor weight gain and vocalization were reported in two
warming, supplemental feeding with artificial bitch’s milk [Esbi- additional puppies. The primary differentials included neona-
lack Pet Ag, Inc.], subcutaneous lactated Ringer’s solution, and tal bacterial sepsis and CHV-1 infection. Because of the lack of
CHAPTER 16  Canine Herpesvirus Infection 169

response to therapy for bacterial infection in the two deceased age and were normal in all. Two of the dogs have become active
puppies, the remaining six puppies were treated with acyclovir guide dogs for the blind, and one is an assistance dog for the
suspension (20 mg/kg PO q6h) for 7 days, and exogenous warm- hearing impaired. All remain physically normal at more than 26
ing adequate to raise the rectal temperature to just above 37.7°C months of age.
(100°F). Therapy was initiated the day before the PCR assay con-
firmed infection with CHV-1. Comments: The epizootiology of this outbreak suggests that
All six treated puppies survived and were successfully the puppies were born from a naïve bitch and exposed to CHV-
weaned between 5 and 6 weeks of age. All were placed in pri- 1 from another carrier dog at days 14 to 17 after birth. Two of 8
vate homes and had uneventful subsequent health histories (25%) succumbed to the disease; 2 of the remaining 6 showed
and normal behavior. Physical examinations including com- early clinical signs, after which antiviral therapy was initiated
plete fundic evaluations were performed at 12 to 14 months of for all dogs.

REFERENCES 8. Burr PD, Campbell ME, Nicolson L, et al. Detection of canine her-
pesvirus 1 in a wide range of tissues using the polymerase chain
1. Carmichael LE, Squire RA, Krook L. Clinical and pathological reaction. Vet Microbiol. 1996;53:227-237.
features of a fatal viral disease of newborn pups. Am J Vet Res. 9. Decaro N, Amorisco F, Desario C, et al. Development and valida-
1965;26:803-814. tion of a real-time PCR assay for specific and sensitive detection of
2. Carmichael LE, Greene CE. Canine herpesvirus infection. In: canid herpesvirus 1. J Virol Methods. 2010;169:176-180.
Greene CE, ed. Clinical Microbiology and Infectious Diseases of 10. Plumb DC, ed. Plumb’s Veterinary Drug Handbook. Pocket Size.
the Dog and Cat. Philadelphia, PA: WB Saunders; 1990:252-258. 6th ed. Hoboken, NJ: Wiley-Blackwell; 2008:18-19.
3. Carmichael LE. Herpesvirus canis: aspects of pathogenesis and 11. Filer CW, Ramji JV, Allen GD, et al. Metabolic and pharmacoki-
immune response. J Am Vet Med Assoc. 1976;156:1714-1725. netic studies following oral administration of famciclovir to the cat
4. Johnson SD, Root Kustritz MV, Olson PN. The neonate—from and dog. Xenobiotica. 1995;25:477-490.
birth to weaning. Canine and Feline Theriogenology. Philadelphia, 12. Malone EK, Ledbetter EC, Rassnick KM, et  al. Disseminated
PA: WB Saunders; 2001:146-167. canine herpesvirus-1 infection in an immunocompromised adult
5. Carmichael L. Neonatal viral infections of pups: canine herpesvirus dog. J Vet Intern Med. 2010;24(4):965-968.
and minute virus of canines (canine parvovirus-1). In: Carmichael 13. Verstegen J, Dhaliwal G, Verstegen-Onclin K. Canine and feline
L, ed. Recent Advances in Canine Infectious Diseases. Ithaca, NY: pregnancy loss due to viral and non-infectious causes: a review.
International Veterinary Information Service (www.Ivis.org); 1999. Theriogenology. 2008;70(3):304-319.
6. Percy DH, Carmichael LE, Albert DM, et al. Lesions in puppies sur-
viving infections with canine herpesvirus. Vet Pathol. 1971;8:37-53.
7. Evermann JF. Diagnosis of canine herpetic infections. In: Kirk RW,
Bonagura JD, eds. Current Veterinary Therapy. Philadelphia, PA:
WB Saunders; 1989:1313-1316.
CHAPTER 17

Canine Viral Respiratory Infections


Jane E. Sykes

after a period of contact with large numbers of other dogs at


Overview of the Canine Viral Respiratory dog parks, dog sporting events such as fly ball, or dog behavior
Infections classes. It can also occur after dogs (or dog owners) visit veteri-
First Described: Respiratory disease in dogs (due to canine nary hospitals, boarding facilities, or pet daycare centers.
adenovirus-2) was first described in 1961 in Canada.1 The With the widespread clinical application of molecular diag-
most newly recognized virus, canine pneumovirus, was nostic assays, it is increasingly apparent that the number of
described in the United States in 2010.2 viruses that can infect the canine respiratory tract is much
larger than previously thought. This has led to exciting new
Proposed Causes: Canine adenovirus, canine influenza virus,
discoveries in the field of canine infectious respiratory disease,
parainfluenza virus, canine respiratory coronavirus, canine
and our knowledge of the pathogens involved continues to
herpesvirus, possibly reoviruses and canine pneumovirus
expand. Co-infections with multiple viruses and bacteria such
Geographic Distribution: Worldwide as Mycoplasma spp., Bordetella bronchiseptica, and Strepto-
Mode of Transmission: Aerosol transmission or close contact, coccus equi subspecies zooepidemicus are common and con-
sometimes fomites tribute to an increased severity of disease.4-6 Viruses believed
to play a role in canine respiratory disease include canine her-
Major Clinical Signs: Harsh or honking cough, serous nasal pesvirus-1 (CHV-1), canine adenovirus-2 (CAV-2), canine dis-
discharge, conjunctivitis, fever (uncomplicated disease). temper virus (CDV), canine parainfluenza virus (CPiV), canine
Fever, lethargy, inappetence, tachypnea, productive pneumovirus, canine respiratory coronavirus (CRCoV), and
cough, mucopurulent nasal and ocular discharge, rarely canine influenza virus (CIV) (Table 17-1). CAV-1 may also
death (complicated disease). play a role when vaccination is not performed or when it is per-
Differential Diagnoses: Upper and lower respiratory diseases formed improperly.7 Reoviruses may also play a role.8 CDV,
such as airway collapse, bordetellosis, Streptococcus equi CHV-1, and CAV are discussed in more detail in Chapters 15,
subsp. zooepidemicus infection, fungal or protozoal pneu- 16, and 18, respectively. The major bacterial causes of canine
monia, respiratory tract neoplasia, airway foreign bodies, transmissible respiratory disease are covered in Part II, Section
chronic bronchitis, eosinophilic bronchopneumopathy 2 of this book (Chapters 34, 38, and 40). Although most of
or granulomatosis, parasitic infections (such as Filaroides, the canine respiratory viruses have a worldwide distribution,
Oslerus, Capillaria, Paragonimus, Dirofilaria), aspiration their relative prevalence varies from year to year and between
bronchopneumonia, left-sided congestive heart failure geographic locations. Even within a state or city, predominant
Human Health Significance: Viruses that cause canine infec- pathogens may differ from one shelter and boarding kennel to
tious respiratory disease generally are not considered another.
zoonotic. Canine parainfluenza and reoviruses repre- CDV, CHV-1, CPiV, CRCoV, and CIV are enveloped
sent potential zoonoses, but the significance of this is viruses, so they survive poorly in the environment and are sus-
unknown. ceptible to a variety of disinfectants. Despite this, contact with
virus that persists in the environment may be important for
transmission in densely housed populations of dogs. CAV-2 is
a non-enveloped virus and has the potential to survive several
weeks on fomites.
Etiology and Epidemiology
Canine Adenovirus-2
Canine viral respiratory disease is a widespread problem where Adenoviruses are icosahedral DNA viruses that infect a vari-
large numbers of dogs are housed indoors together, such as ety of animal species. CAV-2 is found worldwide and primarily
in shelters, commercial dog colonies, and breeding facilities. infects the respiratory tract of dogs. Rarely, it has been impli-
The longer dogs are housed in a shelter situation, the greater cated as a cause of enteritis in dogs, and it was found in the
the risk that respiratory illness will occur.3 In shelter environ- brains of puppies with neurologic signs.9 The virus replicates
ments, transmissible respiratory disease (or canine infectious in nonciliated bronchiolar epithelial cells; epithelial cells of the
respiratory disease complex [CIRDC], previously referred to as nasal mucosa, pharynx, and tonsillar crypts; mucous cells in the
“­kennel cough” or canine infectious tracheobronchitis) delays trachea and bronchi; and type 2 alveolar epithelial cells. CAV-2
the placement of dogs in homes and can result in unmanageable can also be isolated from retropharyngeal and bronchial lymph
costs related to treatment, quarantine, and isolation. Trans- nodes as well as epithelial cells of the intestinal tract. Shedding
missible respiratory disease occasionally occurs in owned dogs typically ceases 1 to 2 weeks after initial infection.

170
CHAPTER 17  Canine Viral Respiratory Infections 171

TABLE 17-1
Potential Causes of Transmissible Respiratory Disease in Dogs
Organism Incubation Period (days)* Shedding Period* Environmental Survival
Canine adenovirus-1 4 to 9 6 to 9 months Weeks to months
Canine adenovirus-2 3 to 6 1 to 2 weeks Weeks to months
Canine distemper virus 3 to 6, longer for neurological signs Weeks to months Hours
Canine herpesvirus-1 6 to 10; may be longer with Unknown Hours
­stress-induced reactivation
Canine influenza virus 2 to 4 7 to 10 days Hours
Canine parainfluenza virus 3 to 10 8 to 10 days Hours
Canine respiratory coronavirus Probably days 6 to 8 days Hours
Bordetella bronchiseptica 2 to 6 Months Has the potential survive and
grow in environmental water
for several weeks
Mycoplasma cynos 3 to 10 Months Hours
Streptococcus equi subspecies Probably days At least 2 weeks Unknown; other streptococci
zooepidemicus can survive weeks

*Incubation and shedding periods are approximate and may differ when co-infections or immunosuppression operate.

Canine Herpesvirus-1 birds and mammals. These rearrangements occur when two
The extent to which CHV-1 plays a role in respiratory disease different viruses simultaneously infect a host, with subsequent
in dogs has been debated.10 Experimental infections of dogs can genetic reassortment. Occasionally, cross-species transmission
result in rhinitis or signs of tracheobronchitis, and intraocular occurs without alteration of the viral genome.
infection results in conjunctivitis and keratitis.11-14 There is evi- In the United States, CIV emerged in racing greyhounds in
dence of widespread exposure to CHV-1 in dogs worldwide,10 and Florida in 2003 and 2004,17 where it caused hemorrhagic pneu-
a substantial proportion of the dog population may be latently monia and a high mortality. However, serologic evidence of
infected. Like other herpesviruses, CHV-1 becomes latent in infection in the greyhound dog population dates back to 1999.18
neurologic tissues, with precipitation of virus shedding by stress. Infections spread slowly and were subsequently reported in rac-
Consistent with the time delay required for reactivation after ing greyhounds and non-greyhounds in at least 38 U.S. states.
stress, CHV-1 was most frequently detected 3 to 4 weeks after Outbreaks have continued to occur in shelter situations for nearly
dogs were introduced to a rehoming center, whereas CRCoV and a decade after the virus was discovered. The virus that currently
CPiV were most frequently detected in the first and second week. circulates in the United States is an H3N8 virus that closely
Dogs infected with CHV-1 were more likely to have severe respi- resembles an equine influenza virus, which suggested that an
ratory disease, although severe respiratory disease itself might interspecies jump occurred without genetic reassortment.17
predispose dogs to the shedding of CHV-1.10 Infection with Instead, accumulation of point mutations with minor amino
CHV-1 was reported in association with fatal hepatic necrosis in acid changes occurred, with sustained transmission among
an adult dog that lacked any evidence of immunosuppression.15 dogs. The most significant outbreaks of disease due to CIV have
Refer to Chapter 16 for information on CHV-1 infections in occurred in Florida, New England, Colorado, Wyoming, and
neonates. Texas. In many other states, sustained transmission of the virus
from one dog to another has not occurred. The most significant
Canine Influenza Virus risk factor for infection has been indoor housing.19 Virtually all
Influenza viruses are enveloped viruses with segmented single- cases to date have involved dogs in kennels, animal shelters, or
stranded RNA genomes that belong to the family Orthomyxo- dog daycare facilities. Dogs of all ages and breeds are suscep-
viridae. Influenza viruses that cause disease in domestic animals tible, but to date severe hemorrhagic pneumonia has occurred
belong to the genus Influenzavirus A, whereas influenza B and only in greyhounds.20 The virus is shed for up to 7 to 10 days.
influenza C viruses primarily circulate among humans. Influenza CIV has retained the ability to infect horses, but horses develop
A viruses are classified based on the genetic composition of their only mild disease or no clinical signs.20
hemagglutinin (H) and neuraminidase (N) genes. To date, 16 Avian-lineage H3N2 CIV emerged in South Korean dogs
H types and 9 N types have been identified, each of which are in 2007, and a similar virus was subsequently isolated from
antigenically distinct.16 The names of influenza viruses are spec- dogs in China.21,22 Experimentally, cats are also susceptible
ified as follows: influenza genus (A, B, or C)/host/geographic to infection by this virus.23 Korean isolates were associated
origin/strain number/year of isolation and, in parentheses, H with epidemics of respiratory disease in kennels within veteri-
and N type—for example, A/canine/Florida/43/2004 (H3N8).17 nary clinics.22 A novel H3N1 virus was also detected in South
Extensive genomic rearrangements that occur within influenza Korean dogs that lacked signs of respiratory disease.24 A novel
A viruses allow for occasional cross-species transmission among H5N2 influenza virus was recently detected in a dog with
172 SECTION 1  Viral Diseases

respiratory disease in China.25 Dogs are susceptible to infection


with human influenza virus H1N126,27 and avian H5N1,28 but BOX 17-1
sustained transmission of these viruses in the dog population
has not been reported. Limited infection of dogs with equine
Genetic Relationships between Coronaviruses
H3N8 viruses was detected in hounds in England29 and during
an equine influenza outbreak in Australia.30 In England, disease Group 1a
was so severe that several hounds had to be euthanized, and Feline coronavirus
subacute bronchointerstitial pneumonia was detected at nec- Canine enteric coronavirus
ropsy.29 The Australian dogs developed inappetence, lethargy, Transmissible gastroenteritis virus of swine
nasal discharge, and a cough that persisted for several weeks, Porcine respiratory coronavirus
but dog-to-dog transmission was not identified. Experimental Ferret and mink coronaviruses
transmission of H3N8 influenza virus from horses to dogs was
documented in Japan, but infected dogs did not show signs of Group 1b
illness.31 Bat coronavirus HKU8
Human coronavirus 229E
Canine Parainfluenza Viruses Human coronavirus NL63
CDV, CPiV, and canine pneumovirus belong to the family Porcine epidemic diarrhea virus
­Paramyxoviridae, which are enveloped RNA viruses. CPiV
belongs to the genus Rubulavirus. Although previously referred Group 2a
to as canine parainfluenza virus-2, it is probably the same virus Canine respiratory coronavirus
as simian virus 5, which was originally isolated from monkey Bovine coronavirus
cell cultures. It has been proposed that it be renamed parainflu- Human coronavirus OC43
enza virus 5.32 Unlike CDV, the outer envelope possesses not Human coronavirus HKU3
only hemagglutinin but also neuraminidase activity (HN attach- Mouse hepatitis virus
ment glycoprotein). The virus infects dogs worldwide, replicates Porcine hemagglutinating encephalomyelitis virus
in epithelial cells of the upper respiratory tract, and often causes
no signs or mild respiratory illness. Respiratory disease may be Group 2b
more severe when co-infections with other pathogens such as SARS coronavirus (humans)
B. bronchiseptica are present.33 Viremia seems to be uncom- Bat SARS coronavirus HKU3
mon, but occasionally CPiV has been isolated from the liver,
spleen, and kidneys. A strain of CPiV was also isolated from a Group 3
dog with neurologic signs.34 There is some evidence that cats Avian infectious bronchitis virus
may be infected with CPiV or a closely related virus.35 Virus is Turkey coronavirus
shed for up to 10 days after infection.
Canine pneumovirus belongs to the genus Pneumovirus. It SARS, Severe acute respiratory syndrome
was first isolated from dogs with acute respiratory disease in
shelters in the United States in 2010.2 The virus is most closely
related to a murine pneumovirus and in fact can replicate in group 2a coronavirus (family Coronaviridae, genus ­Coronavirus)
mice and cause severe inflammatory pathology.36 and is distinct from canine enteric coronavirus, a group 1a coro-
navirus (Box 17-1).39 Minimal serologic cross-reactivity exists
Canine Reoviruses between these two viruses. Group 2a coronaviruses possess a
Reoviruses (family Reoviridae, genus Orthoreovirus) are non- gene that encodes hemagglutinin esterase, an outer membrane
enveloped viruses with a segmented, double-stranded RNA protein glycoprotein. This gene is absent in group 1 and 3 coro-
genome. Mammalian reoviruses infect a variety of host species naviruses. CRCoV is most closely related to a bovine coronavi-
and have a worldwide distribution. The prefix reo- stands for rus but also resembles human coronavirus OC43.40
respiratory enteric orphan virus, which highlights the tropism of CRCoV was first reported in 2003, in a group of dogs with
reoviruses for cells of the respiratory and gastrointestinal tract, respiratory disease in a rehoming facility in England that had
and their uncommon association with disease. Despite serologic been vaccinated against CAV-2, CDV, and CPiV.6,40 Some of
evidence of widespread exposure to reoviruses in dogs, they the dogs were co-infected with CPiV and CHV-1.6 CRCoV
have been found only rarely in dogs with respiratory disease8,37 spread rapidly and primarily was detected in the first week that
and dogs with enteritis.38 There are three mammalian reovi- dogs were introduced to the kennel, after which time CPiV and
rus (MRV) serotypes, and all three have been detected in dogs. CHV-1 were detected. Alone, CRCoV causes subclinical infec-
The role of reoviruses in disease causation in dogs is unclear, tions or mild respiratory disease, but like human respiratory
because disease has not been reproducible experimentally. It has coronaviruses, it can cause reversible damage to, or loss of, the
been speculated that reoviruses might act synergistically with cilia on respiratory epithelial cells (Figure 17-1). As a result,
other respiratory pathogens to cause disease.10 infected dogs are predisposed to secondary infections. Serologic
evidence of exposure to CRCoV is widespread in dogs from
Canine Respiratory Coronavirus North America, Great Britain and continental Europe, Japan,
Coronaviruses are enveloped RNA viruses that possess large, Korea, and New Zealand,10,41-43 and the virus has been detected
club-shaped spikes on their outer surface, also known as pep- widely using PCR-based methods in dogs with respiratory dis-
lomers (see Figure 14-1, B). CRCoV is a relatively newly identi- ease from many of these countries. To date, there is no evidence
fied cause of contagious respiratory disease in dogs. The virus is a that cats can be infected with CRCoV.
CHAPTER 17  Canine Viral Respiratory Infections 173

A B
A
FIGURE 17-1  Effect of canine respiratory coronavirus infection on ciliated respiratory
epithelial cells. A, Respiratory epithelium immediately before inoculation with CRCoV. B,
The same cells 48 hours after inoculation with CRCoV. There is marked loss of cilia, thinning
of the epithelium with individual cell necrosis, and sloughed necrotic cells on the surface.
There are also occasional large vacuoles within the epithelium, which likely represent foci
of cell loss secondary to viral replication. (Courtesy Dr. Simon Priestnall and Professor Joe
Brownlie, Royal Veterinary College, UK.)

Infection with pancytotropic strains of canine enteric coro-


navirus has recently been associated with severe systemic dis-
ease in puppies from Europe. Affected pups have fever, mucoid
to hemorrhagic enteritis, lymphopenia, neurologic signs such
as seizures, and bronchopneumonia.44,45 Co-infections with
CPV-2 were present in some puppies, but the disease has been
reproduced by experimental infection by canine enteric corona- B
virus alone. Death occurred in some puppies, but others devel- FIGURE 17-2  A, Right eye of a 10-year-old male neutered miniature schnauzer
oped transient gastrointestinal signs and recovered.46 infected with bilateral conjunctivitis and dendritic ulceration associated with canine her-
pesvirus infection. Fluorescein uptake can be seen in a branching pattern at the limbus of
Clinical Features the right eye. B, The same dog 1 month later. Note diffuse stain uptake over at least 25%
of the corneal surface. (Courtesy University of California, Davis Veterinary Ophthalmology
Signs and Their Pathogenesis Service.)
The incubation period for viral respiratory disease is generally
less than 2 weeks and can be as short as 2 to 3 days. Influ-
enza virus infections, in particular, have been associated with mild fever, a paroxysmal harsh or “honking” cough, serous
very short incubation periods.20 Transmission occurs by aero- nasal discharge, and sometimes sneezing, but usually otherwise
sol, but direct contact between dogs and fomite transmission affected dogs are often alert, active, and appetent. The cough
(hands, clothing, contaminated food and water bowls, common may be followed by gagging or retching, which may be followed
hallways and exercise areas) are important routes in popula- by the production of frothy mucus. An altered bark or stridor
tion-dense environments. Cells of the larynx, trachea, bronchi, may occur in dogs that develop laryngitis, tonsillitis, and/or
and sometimes the nasal mucosa, bronchioles, and alveoli are pharyngitis. CAV-2 and CHV-1 may cause conjunctivitis.47 In
infected, and although viral shedding patterns differ between addition, CHV-1 has been associated with ulcerative and non-
pathogens, shedding may occur before the onset of clinical ulcerative keratitis, which may be precipitated by immunosup-
signs. The duration of shedding for CIV and CPiV is very short, pression (Figure 17-2).48-50 Infections with CIV or CDV, or
typically a few days, and in some dogs, shedding has ceased by infections with multiple respiratory pathogens, may be more
the time clinical signs are most apparent. In contrast, CDV can likely to produce systemic signs of fever and lethargy. Dogs that
be shed for weeks (see Chapter 15). develop secondary bacterial infections may show fever, inappe-
Infection with respiratory viruses may be associated with tence, lethargy, a mucopurulent nasal discharge, tachypnea, and
no signs, or complicated pneumonia and death can occur. In a moist, productive cough.
general, morbidity is high, but mortality is low. Moderate to
severe signs may be more likely to occur in very young puppies, Physical Examination Findings
genetically susceptible animals, and when stress and co-infec- On physical examination, dogs with uncomplicated transmissible
tions with multiple viral and bacterial pathogens are present. respiratory disease are typically bright, alert, and active with a
Infection with CDV is especially effective at predisposing dogs paroxysmal cough. The cough is often easily elicited on tracheal
to other respiratory viral infections, because of its immuno- palpation. Conjunctivitis and serous ocular and/or nasal dis-
suppressive properties, but other viruses that damage ciliated charge may be present, and the tonsils may be enlarged and hyper-
epithelial cells, such as CRCoV and CPiV, also predispose to emic. Dogs with secondary bacterial infections may be pyrexic,
co-infections. Clinical signs of acute respiratory disease include lethargic, and tachypneic with increased respiratory effort and
174 SECTION 1  Viral Diseases

TABLE 17-2
Diagnostic Assays Available for Respiratory Viruses in Dogs
Assay Specimen Type Target Performance
Virus isolation Conjunctival, nasal, and caudal Virus False negatives can occur in specimens that contain
pharyngeal swabs; transtracheal no or low numbers of virus particles. Attenuated
and bronchoalveolar wash speci- live vaccine virus may also be isolated. May take
mens; airway and lung speci- several days and requires specialized techniques
mens collected at necropsy and expertise.
Influenza ELISA Same as virus isolation CIV antigen Inexpensive and rapid. Sensitivity and specific-
antigen assay ity in naturally infected dogs have not been well
established, so results should be interpreted with
caution.
PCR or RT-PCR Same as virus isolation Viral nucleic acid Sensitivity and specificity may vary with assay design.
Sensitivity may be low because of brief shedding
for some viruses. Assays may not be available for
all viruses or may not detect all virus strains. Test-
ing specimens from multiple different anatomic
sites or combining PCR with other diagnostic tests
can increase sensitivity. Attenuated live vaccine
virus may be detected after vaccination. Because of
subclinical shedding, the significance of a positive
result may be difficult to interpret. False-negatives
may occur as a result from degradation of viral
nucleic acid during specimen transport.
Serology Serum Antibodies against Interpretation complicated by previous vaccination
respiratory viral and exposure. May be useful when the exposed
antigens population is naïve and unvaccinated for the virus
of interest. Acute and convalescent sera required
for diagnosis.
Histopathology Necropsy specimens Viral inclusions; Can be used for diagnosis at necropsy. Detection of
immunostaining inclusions has low sensitivity, and immunostaining
and/or PCR is required to definitively identify the
infecting virus.

CIV, Canine influenza virus; RT, reverse transcriptase.

increased lung sounds on thoracic auscultation. A mucopurulent situation can facilitate diagnosis and allow interpretation of the
nasal discharge may also be present. Ulcerative or nonulcerative significance of positive test results. Organism detection meth-
keratitis may be present in dogs infected with CHV-1. ods, such as PCR assays, are likely to be of highest yield early
in the course of illness (e.g., the first 1 to 3 days) or in exposed
Diagnosis dogs that have not yet developed clinical signs. Use of a combi-
nation of serology and organism detection methods also facili-
It is not possible to identify the cause of transmissible respi- tates diagnosis. In shelter situations or outbreaks where severe
ratory disease in dogs based on clinical signs alone, because disease occurs, necropsies can provide valuable information and
each pathogen produces a similar spectrum of signs. The high should be performed by a veterinary pathologist as soon as pos-
prevalence of co-infections further complicates diagnosis. In sible after death or euthanasia. Tissues should be submitted for
addition, other, noncontagious causes of respiratory disease in histopathology (in formalin), bacterial and virus cultures (fresh
dogs can lead to signs that closely resemble transmissible respi- tissue), and PCR assay for respiratory viruses and bacteria (fresh
ratory disease. A history of exposure to other dogs can support or frozen tissue; see Chapter 5).
the diagnosis.
Most dogs experience self-limiting disease. Attempts to obtain Laboratory Abnormalities
an etiologic diagnosis should be made when disease persists for There are no specific abnormalities in the CBC, serum biochem-
longer than 7 to 10 days or is complicated by bacterial pneumo- istry profile, or urinalysis that aid in a diagnosis of canine viral
nia, with lethargy and inappetence. When outbreaks occur in respiratory disease. The CBC may be normal or show a mild to
shelters or the pattern of endemic respiratory disease changes, moderate neutrophilia. Band neutrophils and neutrophil toxic-
attempts to make an etiologic diagnosis are also indicated and ity may be apparent in dogs with secondary bacterial pneumo-
encouraged (Table 17-2). Collection of multiple specimen types nia. Severe infections may be associated with leukopenia, which
from several dogs with and without clinical signs in an outbreak may be followed by leukocytosis in dogs that recover.
CHAPTER 17  Canine Viral Respiratory Infections 175

Microbiologic Tests
Virus Isolation
Efforts to isolate viruses from dogs with acute respiratory
disease have been most useful for identification of novel or
reemerging pathogens.2,38,40 Despite the increased availability
of molecular diagnostic assays, virus isolation is still offered to
veterinarians for routine diagnostic purposes by laboratories
that specialize in virology (e.g., the Animal Health Diagnos-
tic Laboratory at Cornell University in Ithaca, NY). Suitable
specimens for respiratory virus isolation are nasal and pharyn-
geal swab specimens, transtracheal or bronchoalveolar lavage
specimens, or upper airway or lung tissue obtained at nec-
ropsy. If swabs are used to collect specimens, polyester-tipped
swabs and specific virus transport media should be used. Cot-
ton swabs should be avoided, because influenza viruses adhere
to the cotton, which can lead to reduced sensitivity.20 Virus
isolation is a specialized process that may take several days.
As with PCR assays, sensitivity may be low because shedding
of respiratory viruses occurs early in the course of illness and
may be transient. CRCoV fails to replicate in many cell lines,
but was successfully isolated in HRT-18 cells.51 This may
explain its relatively recent discovery relative to other respira-
tory viral pathogens.

Fluorescent Antibody Testing


Fluorescent antibody can be applied to smears made from swabs
from the upper respiratory tract, or tissues collected at necropsy,
such as fresh lung tissue. Assays are available for detection of
CPiV, CAV, CDV (see Chapter 15), and CHV-1. Attenuated
FIGURE 17-3  Dorsoventral thoracic radiograph from a 2-year-old male neutered live vaccination with intranasal vaccines for CPiV and CAV has
coonhound that had left a boarding facility 6 days previously with a moist, productive the potential to interfere with fluorescent antibody results, but
cough that subsequently progressed and was complicated by the development of fever this requires further study. The sensitivity of fluorescent anti-
and a mucopurulent nasal discharge. A CBC showed neutrophilia (17,840 cells/µL), body testing is likely to be lower than that of PCR assays, and
increased numbers of band neutrophils (223 cells/µL), and monocytosis (3,345 cells/µL). inexperienced personnel may misinterpret nonspecific fluores-
Bronchopneumonia is present that involves the right cranial, right middle, and left cranial
cence as a positive result.
lung lobes. Interestingly, canine herpesvirus DNA was detected in a pooled extract from
nasal, conjunctival, and oral swab specimens, as well as a whole blood sample using a
PCR assay. Enzyme-Linked Immunosorbent Assays for Influenza Virus Antigen
Point-of-care ELISAs are available for detection of nucleopro-
tein antigen of human influenza A viruses. The assays are easily
performed and provide rapid results. Unfortunately, such assays
have had limited sensitivity and specificity for diagnosis of CIV
Transtracheal wash and bronchoalveolar lavage specimens infections, and so their use is not recommended. False positives
from dogs with viral pneumonia typically show a suppurative or may especially be a problem in shelters where canine influenza
mixed exudate, sometimes with evidence of intracellular bacteria. is not endemic.
Culture for aerobic bacterial culture and Mycoplasma spp. are
indicated on wash specimens, and antimicrobial susceptibilities Molecular Diagnosis Using the Polymerase Chain Reaction
should be obtained for any aerobic bacteria isolated. Organisms Panels of real-time PCR assays that detect respiratory patho-
such as Pasteurella spp., Staphylococcus pseudintermedius, Strep- gens are offered by some commercial veterinary diagnostic
tococcus canis, Escherichia coli, Klebsiella pneumoniae, and some laboratories. These may include assays for CRCoV, parainflu-
Mycoplasma spp. infect the airways as opportunists and are not enza virus, and CIV, as well as bacterial pathogens such as B.
considered to be primary pathogens. Bacterial culture of nasal bronchiseptica and Mycoplasma spp. Swabs of the nasal cav-
swabs is generally not recommended because it often leads to ity and/or caudal pharynx, or respiratory lavage specimens
growth of normal flora, which has no clinical relevance. could be submitted for testing. At necropsy, CRCoV is most
readily detected in upper respiratory tract specimens, such as
Diagnostic Imaging the nasal mucosa, nasal tonsil, and trachea.52 Lung specimens
Plain Radiography may also yield positive results. Unfortunately, false-negative
In uncomplicated viral infections, plain thoracic radiography PCR results are common, especially in antemortem specimens,
in dogs often shows no significant abnormalities, or there may because of transient or low-level shedding of many respiratory
be a mild diffuse interstitial or bronchointerstitial pattern. Sec- viruses. In addition, because many respiratory viruses are RNA
ondary bacterial bronchopneumonia may be characterized by viruses and RNA is very labile when compared with DNA, false
development of peribronchial and alveolar infiltrates or lobar negatives can occur when viral RNA degrades during specimen
consolidation (Figure 17-3). transport.
176 SECTION 1  Viral Diseases

In outbreak situations, false-positive PCR results may occur also been described. Hemorrhagic interstitial and bronchoint-
if swab specimens become contaminated with virus from the erstitial pneumonia, vasculitis, and thrombus formation have
environment or the hands of personnel. Clean examination been observed in greyhounds with CIV infection.20 CAV-2 can
gloves should be worn for each dog, and the swab should only produce large, basophilic intranuclear inclusion bodies within
touch the anatomic site to be tested. Vaccination with live bronchial, bronchiolar, and alveolar cells. These must be differ-
attenuated vaccines (especially intranasal vaccines) may cause entiated from the eosinophilic inclusions of CDV (see Chapter
positive test results with real-time PCR assays, but the extent to 15).55 Reoviruses can also produce intracytoplasmic inclusions
which this occurs in dogs with respiratory viral illness requires within bronchial epithelial cells.8 Use of immunohistochemistry
further study. Because CIV vaccines are parenteral inactivated can facilitate definitive identification of viral antigen within cells
vaccines, they would not be expected to lead to false-positive of the respiratory tract.5,55
PCR assay results. The detection of nucleic acid from a respira-
tory virus in a specimen may not imply disease causation. Treatment and Prognosis
Serologic Diagnosis Like the common cold in humans, contagious respiratory dis-
Diagnostic laboratories that specialize in veterinary virology ease in dogs resolves without treatment in the vast majority of
may offer serologic assays for antibodies to canine respiratory dogs, regardless of the underlying cause. For dogs with signs
viral pathogens on a commercial basis to practitioners. Serology of respiratory disease that have been present for less than 7 to
has limited use for diagnosis because of vaccine titer interference 10 days and that remain bright and appetent, no treatment is
for some organisms, and the high prevalence of subclinical expo- indicated. In some dogs, cough persists for as long as 10 to 30
sure to organisms endemic in the dog population. Titers may days. Cough suppressants such as hydrocodone could be used for
be negative in the first 10 days of illness, and some dogs may dogs with a nonproductive, honking cough that occurs through-
not show a significant increase in antibody titer after infection. out the day and night. Cough suppressants should not be used in
Despite these limitations, serologic assays have been key to iden- dogs that have productive cough, because they suppress normal
tification of infection and disease caused by emerging pathogens clearing mechanisms. Use of a harness or gentle leader for leash
such as CIV, when the disease is not endemic.20 In this situation, walking rather than a neck collar may also reduce cough. The
analysis of paired serum specimens collected 2 weeks apart can efficacy and optimal dosage of neuraminidase inhibitors such
be used to document recent infection. In some dogs, no other as oseltamivir (see Chapter 7) is unknown, and because of this
diagnostic test may be useful for antemortem diagnosis because and the fact that oseltamivir is a first-line treatment for pandemic
virus shedding is so transient and difficult to detect. influenza in humans, it should not be used to treat dogs with
Serologic assays for CIV exposure are based on serum neu- respiratory disease, even when CIV infection is present. For dogs
tralization or hemagglutination-inhibition (see Chapter 2). with confirmed ocular CHV-1 infections that are associated with
Assays for CIV that use equine influenza virus antigen for corneal ulceration, topical antiviral ophthalmic preparations such
antibody detection have suboptimal sensitivity, whereas an as idoxuridine or cidofovir could be considered (see Chapter 7).50
assay based on H3N8 CIV antigen was sensitive and specific Antimicrobial drug treatment could be considered for dogs
when compared with serum neutralization.53 Seroconver- with signs that persist beyond 7 to 10 days, but is primarily
sion to CRCoV in a bovine coronavirus antigen ELISA assay indicated when there is evidence of secondary bacterial bron-
helped identify the association between infection with this chopneumonia, such as pulmonary alveolar infiltrates and con-
virus and respiratory disease in dogs in the rehoming kennel solidation on thoracic radiography, lethargy, mucopurulent
in the United Kingdom.6 Serum neutralization assays can be oculonasal discharges, and/or decreased appetite. Because anti-
performed for detection of antibodies to CAV-2, CPiV, and microbial drug resistance is increasingly reported among sec-
CHV-1. Antibodies to CAV-2 and CPiV can also be detected by ondary bacterial pathogens and B. bronchiseptica, treatment of
hemagglutination-inhibition. dogs with secondary bacterial bronchopneumonia is optimally
based on the results of culture of a transtracheal wash specimen
Pathologic Findings from each affected dog and antimicrobial susceptibility test-
Gross Pathologic Findings ing. For dogs with severe pneumonia, initial treatment should
Necropsy of dogs with viral respiratory infections may show involve the use of broad-spectrum parenteral antimicrobial
no gross lesions, or pulmonary consolidation and hyperemia of drugs such as a combination of a fluoroquinolone and a peni-
the tracheal mucosa may be present. Purulent exudate may be cillin or clindamycin. When infection with B. bronchiseptica
present in the bronchi. Greyhounds that died of CIV infection or Mycoplasma spp. is suspected, doxycycline may be the best
had extensive hemorrhage within the lungs, mediastinum, and first choice. Indiscriminate use of antimicrobial drugs leads only
pleural space, together with mild, fibrinous pleuritis.17 to widespread bacterial resistance and failure of antimicrobial
drug treatment in dogs that develop severe disease.
Histopathologic Findings Dogs with pneumonia may also require treatment with intra-
Histopathology of the airways of dogs with viral respiratory venous fluids, supplemental oxygen, nebulization, and coup-
infections may show loss or irregularity of normal respiratory age. Nutritional support in the form of feeding tubes may be
cilia and epithelial necrosis and ulceration within the trachea, required for dogs that are inappetent.
bronchi, and bronchioles. With secondary bacterial infection, Prognosis depends on the virulence of the causative agent(s),
a neutrophilic or mixed inflammatory infiltrate may be seen the presence of co-infections, and other factors that contribute
in the airway mucosa, submucosa, and alveoli.54 Bronchial to host immunosuppression. Prognosis is generally excellent for
and bronchiolar lumina may contain neutrophils, macro- dogs with uncomplicated infections with a single pathogen. For
phages, and cellular debris. Thickening of the alveolar walls CIV infection, mortality rates have been less than 8% and could
with type 2 pneumocyte hyperplasia and interstitial edema has be lower with rapid diagnosis and appropriate treatment.20
CHAPTER 17  Canine Viral Respiratory Infections 177

Immunity and Vaccination in antibody titers that persisted for at least 2 years.62 Use of
intranasal CPiV vaccines, as opposed to parenteral vaccines, has
Vaccines are available for reduction of disease due to CPiV, been advocated, because they produce local immunity, reduce
CAV-2, CDV, and CIV. With the exception of CDV vaccines, shedding, and can be used in puppies as young as 3 to 4 weeks
none of the available vaccines completely prevent infection and of age.63 However, proper administration of mucosal vaccines
shedding, but they can lessen the severity of disease, provided may be difficult in aggressive dogs or dogs that refuse to be
other factors such as overcrowding and appropriate disinfec- restrained. Mucosal vaccines may also be associated with tran-
tion and reduction of other stressors are also addressed. Paren- sient respiratory illness for 3 to 10 days after immunization in
teral and mucosal (intranasal and oral) attenuated live vaccines a small percentage of dogs. This may be problematic in shelter
are available for CPiV and CAV-2. The vaccine for CIV is an and boarding environments, because these signs cannot be dis-
inactivated vaccine. Vaccines for canine transmissible respira- tinguished from those that result from natural infection with
tory disease are considered noncore vaccines, so they should wild-type viruses. More studies that evaluate the relative effi-
be administered to dogs at risk of exposure, such as those that cacy of mucosal and parenteral CPiV vaccines are required.
enter shelters, boarding kennels, shows, sporting competitions,
popular dog parks, or pet daycare facilities. Prevention
Canine Adenovirus-2 Although vaccination can reduce the prevalence of respira-
Mucosal and parenteral vaccines are available for prevention tory disease in dogs,3 current vaccines do not provide protec-
of disease due to CAV-2. Parenteral CAV-2 vaccines also pro- tion against all the organisms that cause respiratory disease in
tect against CAV-1 infection. Maternal antibodies persist for up dogs, and immunity is not sterile, so infection and mild clini-
to 12 to 14 weeks after birth. Mucosal vaccines may be useful cal signs can still occur. When factors such as stress, immuno-
to overcome maternal antibodies in young dogs that are intro- suppression, co-infections, and overwhelming challenge doses
duced to shelter environments. However, parenteral vaccines are present, vaccine-induced protection may be overwhelmed.
are still required for adequate protection against CAV-1. Prevention of transmissible respiratory tract disease in dogs
therefore involves not only vaccination, but also control meth-
Canine Herpesvirus ods that include quarantine of dogs introduced into densely
An inactivated vaccine for CHV-1 is available in Europe for populated environments, early identification and isolation of
pregnant bitches to protect puppies against neonatal infections dogs with signs of respiratory disease through proper training
(see Chapter 16). It is not intended to reduce respiratory disease of shelter personnel, avoidance of overcrowding and mixing of
due to CHV-1 infection. dogs, use of solid walls between runs, reduction of time dogs
spend in a shelter environment, optimum nutrition, fomite con-
Canine Influenza Virus trol, control of noise such as barking, and proper ventilation
Inactivated, parenteral vaccines are available for reduction of and disinfection (see Box 11-5). Distances traveled by aerosols
disease and shedding caused by H3N8 CIV.56 One vaccine also generated by dogs are unknown. Dogs with respiratory disease
reduced the severity of illness caused by co-challenge with CIV should be separated from other dogs in shelter environments
and Streptococcus equi subspecies zooepidemicus.4 The use of by at least 25 feet (or as far as possible) and ideally placed in
these vaccines could be considered for dogs that are likely to an enclosed room with a separate ventilation system. Isolated
contact other dogs in regions where CIV is endemic, especially dogs should not be moved back into the general shelter popula-
those that enter boarding or pet daycare facilities. Vaccination tion, but adopted out of isolation or a separate recovery room.
against CIV is required for importation of North American dogs Provided proper cleaning and contact times are used, use of dis-
to Australia.57 The initial vaccine can be given as early as 6 infectants with activity against parvovirus should also kill respi-
weeks of age. Because CIV vaccines are inactivated, two initial ratory viruses, with CAV-2 being the respiratory virus that is
doses are required 3 to 4 weeks apart, and maximum immunity most resistant to disinfection. The order of inspection of dogs
does not occur until 1 week after the second dose. As a result, in shelters should be healthy dogs, then quarantined dogs, and
CIV vaccines may not protect dogs that enter shelters where finally dogs in isolation.
canine influenza is endemic, unless newly introduced dogs are When outbreaks occur, attempts should be made to iden-
separated from dogs that might be shedding CIV until immuni- tify the pathogen(s) involved. This allows potential routes of
zation is complete. Annual boosters are recommended for dogs transmission of infection and shedding patterns to be identified,
that remain at risk of infection, but the maximum duration of proper vaccination and control strategies to be implemented,
immunity is unknown. and adequate disinfectants to be selected. For example, quar-
antine for 2 weeks and isolation or removal of any dogs that
Canine Parainfluenza Virus develop illness may be effective for diseases such as canine influ-
Intranasal vaccination with CPiV significantly reduces clinical enza, because of the short incubation and shedding periods.
signs and virus shedding after challenge.58-60 One study showed However, it is less likely to be successful for distemper and bor-
that intranasal CPiV vaccination reduced clinical signs even detellosis, which can have prolonged incubation and shedding
when dogs were challenged as long as 1 year after immuniza- periods. For some viral respiratory diseases such as canine influ-
tion.58 Few studies have compared the efficacy of parenteral enza, discontinuation of intake and adoptions for 2 to 3 weeks
and intranasal vaccines. In a study reported in the early 1980s, may be necessary while the disease runs its course. Exposed
the parenteral vaccine was less effective at reducing shedding dogs could also be transferred to foster care in a household with
than the intranasal vaccine,59 but a study reported in the 1970s no other dogs. Because of a lack of adequate resources for treat-
showed reduction of clinical signs and shedding after parenteral ment, diagnostic evaluation, and management, some shelters
vaccination.61 The use of one parenteral CPiV vaccine resulted have elected to euthanize large numbers of affected dogs.
178 SECTION 1  Viral Diseases

Shelters should provide written information on transmis- Public Health Aspects


sible respiratory disease to clients who adopt shelter animals,
regardless of the presence or absence of clinical signs at the time Viruses related to simian virus 5 (parainfluenza virus 5) have
of adoption, and communicate fully about diseases that are been detected in humans, but the role of CPiV as a human
endemic in the shelter. Whenever possible, dogs that leave shel- pathogen is controversial. Reoviruses have a broad host range,
ters should be kept away from other dogs for at least 2 weeks and human infection has been associated with enteritis and
(consider 4 weeks if bordetellosis is endemic), and informed respiratory disease; MRV-3 infection was associated with men-
about follow-up medical care. Other dogs in the household ingitis in a child.64 Thus they represent a potential zoonosis.
should be vaccinated before they are placed in contact with the Although adenoviruses, influenza viruses, coronaviruses, and
shelter animal. Clients should also be instructed to ensure their paramyxoviruses cause respiratory disease in humans, there is
local veterinary clinic is aware that their dog is newly adopted no evidence that CAV-2, CIV, CDV, CHV-1, or CRCoV can be
from a shelter, so that precautions can be taken to prevent con- transmitted to humans. Human influenza viruses such as H1N1
tamination of the hospital environment when the dog is exam- can cause disease in dogs and may have the potential to spread
ined a few days after leaving the shelter. from infected dogs back to humans.

CASE EXAMPLE All Other Systems: No abnormalities were detected.


Ophthalmologic Examination: Pupillary light reflexes
(PLRs) (direct and consensual) were brisk and complete. There
Signalment: “Winnie”, a 10-year old male neutered miniature was no evidence of anisocoria. Menace response, dazzle,
Schnauzer from Sacramento, CA and palpebral reflexes were all complete. Globe position
History: Winnie was brought to his local veterinarian be- and movements were normal bilaterally (OU). Periorbital
cause of a 1-day history of serous ocular and nasal dis- palpation and globe retropulsion were also normal. The dog
charge, and sneezing. Fever (103.5°F or 39.7°C) was behaved as if sighted. The eyelids were normal. Both nictitans
documented on physical examination, and enrofloxacin were hyperemic and edematous, and there was moderate
(2 mg/kg PO q24h), bacitracin-neomycin-polymyxin oph- conjunctival hyperemia, chemosis, and mucoid ocular
thalmic ointment (both eyes q8h), and diphenhydramine discharge OU. Bilateral iris atrophy and lens nuclear sclerosis
(2.2 mg/kg PO q12h) were prescribed, without clinical im- were also present. The cornea, anterior chamber, vitreous,
provement. A day later, he became lethargic, inappetent, and dilated fundic examination were normal OU. In the left
and polydipsic, and the owners described continuous eye (OS), a pinpoint incipient anterior cortical cataract was
nasal discharge, and a moist cough. Winnie was brought identified just ventral to the center of the lens. In the right
to the UC Davis VMTH for a second opinion. He had been eye (OD), a linear pigment deposition on the anterior lens
vaccinated regularly for CDV, CAV-2, CPV, rabies, Borde- capsule was present at the 5 o’clock position.
tella bronchiseptica, and CPiV. He was recently cared for in Schirmer Tear Test: 8 mm/min OU
a boarding facility for a week and had been home for the Intraocular Pressure: 16 mm Hg OS, 17 mm Hg OD
past 11 days. He also regularly visited dog parks. There Fluorescein Stain: Multiple dendritic ulcers were identified at
were two other miniature Schnauzers at home, and both the ventral peripheral cornea OD (see Figure 17-2).
were currently well. Laboratory Findings:
Current Medications: Enrofloxacin, 2 mg/kg PO q24h CBC:
Other Medical History: Increased activity of serum ALP was HCT 53.5% (40-55%)
noted at a senior care visit 2 months before the onset of MCV 67.7 fL (65-75 fL)
respiratory signs. MCHC 35.3 g/dL (33-36 g/dL)
Physical Examination: 1 nucleated red cell/100 WBCs
Body Weight: 9.4 kg WBC 14,600 cells/µL (6000-13,000 cells/µL)
General: Lethargic but hydrated. T = 100.8°F (38.2°C), Neutrophils 10,804 cells/µL (3000-10,500 cells/µL)
HR = 104 beats/min, respiratory rate = 12 breaths/min, Band neutrophils 438 cells/µL
mucous membranes pink, CRT = 1 to 2 s. Lymphocytes 1752 cells/µL (1000-4000 cells/µL)
Eyes, Ears, Nose, and Throat: Severe chemosis, hyperemia, Monocytes 1460 cells/µL (150-1200 cells/µL)
and mucoid ocular discharge were present bilaterally. Eosinophils 0 cells/µL (0-1500 cells/µL)
There was also profuse bilateral serous nasal discharge and Platelets 216,000 platelets/µL (150,000-400,000 platelets/µL)
decreased nasal airflow, hypersalivation, and right tonsillar A few highly reactive lymphocytes were noted.
enlargement. Serum Chemistry Profile:
Musculoskeletal: Body condition score 5/9. Sodium 142 mmol/L (145-154 mmol/L)
Respiratory: Stertorous respiratory noises were noted. Potassium 6.1 mmol/L (3.6-5.3 mmol/L)
Increased respiratory effort with a normal respiratory rate Chloride 102 mmol/L (105-116 mmol/L)
was also present. Auscultation revealed referred wheezing Bicarbonate 25 mmol/L (16-26 mmol/L)
noises in all lung fields. Phosphorus 6.7 mg/dL (3.0-6.2 mg/dL)
Gastrointestinal: Tense abdomen, hepatomegaly was Calcium 9.5 mg/dL (9.9-11.4 mg/dl)
detected on abdominal palpation. BUN 20 mg/dL (8-31 mg/dL)
Creatinine 0.7 mg/dL (0.5-1.6 mg/dL)
CHAPTER 17  Canine Viral Respiratory Infections 179

Glucose 80 mg/dL (60-104 mg/dL) to administer topical idoxuridine (0.1% ophthalmic solution,
Total protein 5.7 g/dL (5.4-7.4 g/dL) 2 drops OU q4-6h). Unfortunately a caretaker of the dog
Albumin 2.2 g/dL (2.9-4.2 g/dL) did not regularly administer the idoxuridine. At a recheck 5
Globulin 3.5 g/dL (2.3-4.4 g/dL) weeks after the dog was first seen, the owner reported 80%
ALT 564 U/L (19-67 U/L) improvement in Winnie’s respiratory signs, but he continued
AST 116 U/L (21-54 U/L) to sneeze approximately six times a day and was pawing at
ALP 1024 U/L (15-127 U/L) his eyes. Examination revealed persistent conjunctivitis, and
Gamma GT 10 U/L (0-6 U/L) a fluorescein stain showed large, superficial areas of uptake
Cholesterol 195 mg/dL (135-345 mg/dL) over the central cornea OU (see Figure 17-2). A Schirmer
Total bilirubin 0.4 mg/dL (0-0.4 mg/dL). tear test (STT) was 13 mm/min OS and 8 mm/min OD. A CBC
Urinalysis: SGr 1.009; pH 8.5, negative for protein, bilirubin, was normal, and the serum biochemistry profile showed
hemoprotein, glucose; rare WBC/HPF, 0 RBC/HPF, 0-3 hyaline persistently increased ALP (835 U/L) and GGT activities
casts/HPF, few amorphous crystals, moderate degenerated (12 U/L) and improvement in the serum ALT activity (124
cells and amorphous debris in the sediment. U/L). The owner was instructed to administer idoxuridine
Imaging Findings: for an additional 2 weeks, as well as lubricating ointment,
Thoracic and Cervical Radiographs: A mild to moderate bacitracin-neomycin ophthalmic ointment (q8h OU), and l-
bronchial and interstitial pattern was identified and lysine gel (2 mL [400 mg] q12h PO), separating the ophthalmic
was most prominent in the caudal lung fields. Cervical medications with administration of the idoxuridine first. At a
radiographs were unremarkable. The pulmonary changes recheck 2 weeks later, all signs had resolved and there was
were considered nonspecific, but more significant than no fluorescein uptake. STT results were 14 mm/min OS and
those expected due to age. 10 mm/min OD. The idoxuridine was discontinued. Winnie
Abdominal Ultrasound: The cranial pole of the right adrenal had another episode of respiratory signs 4 months later, at
was mildly to moderately enlarged. The liver appeared which time the STT results were 15 mm/min OU. Treatment
sonographically normal. A 3 mm stone was identified in the with lubricating eye ointment and l-lysine was continued.
urinary bladder. Comments: This was an unusual case of ocular and respiratory
Microbiologic Testing: A nasal swab specimen was herpesviral infection. The early detection of dendritic ulcers
submitted for real-time PCR panel for canine respiratory allowed a connection to be made between the clinical
pathogens, which included CHV-1, CIV, CDV, CPiV, CAV-2, signs and positive PCR assay result. The negative PCR assay
and B. bronchiseptica. The results were available 3 days later, results for other respiratory pathogens did not rule out
and the specimen was positive for CHV-1 DNA. the possibility of co-infections, because shedding of these
Diagnosis: Upper respiratory disease and keratitis associated organisms may have ceased or been at undetectable levels.
with CHV-1 infection. Although idoxuridine was used to treat this dog, topical
Treatment: Winnie was hospitalized in isolation and treated cidofovir may also have been effective.50 Approximately 2
with 0.9% NaCl (30 mL/hr IV), nebulization for 15 min years later, mild serous nasal discharge and depigmentation
q8h, clavulanic acid-amoxicillin (13.5 mg/kg PO q12h), of the planum nasale developed. A biopsy of the nasal
and lubricating eye ointment (1 inch OU q4h). Clinical planum showed epitheliotropic lymphoma. The dog was
improvement occurred a day later, electrolyte abnormalities treated with multiagent chemotherapy for over a year
normalized, and the dog was discharged with instructions without relapse of herpetic keratitis.

SUGGESTED READINGS 4. Larson LJ, Henningson J, Sharp P, et  al. Efficacy of the canine
influenza virus H3N8 vaccine to decrease severity of clinical disease
Buonavoglia C, Martella V. Canine respiratory viruses. Vet Res. after cochallenge with canine influenza virus and Streptococcus equi
2007;38:355-373. subsp. zooepidemicus. Clin Vaccine Immunol. 2011;18:559-564.
Crawford PC, Dubovi EJ, Castleman WL, et al. Transmission of equine 5. Damian M, Morales E, Salas G, et  al. Immunohistochemical
influenza virus to dogs. Science. 2005;310:482-485. detection of antigens of distemper, adenovirus and parainflu-
Ellis JA, Krakowka GS. A review of canine parainfluenza virus infection enza viruses in domestic dogs with pneumonia. J Comp Pathol.
in dogs. J Am Vet Med Assoc. 2012;240:273-284. 2005;133:289-293.
Erles K, Dubovi EJ, Brooks HW, et  al. Longitudinal study of viruses 6. Erles K, Dubovi EJ, Brooks HW, et  al. Longitudinal study of
associated with canine infectious respiratory disease. J Clin Micro- viruses associated with canine infectious respiratory disease. J Clin
biol. 2004;42:4524-4529. Microbiol. 2004;42:4524-4529.
7. Decaro N, Campolo M, Elia G, et al. Infectious canine hepatitis: an
REFERENCES “old” disease reemerging in Italy. Res Vet Sci. 2007;83:269-273.
1. Ditchfield J, Macpherson LW, Zbitnew A. Association of canine 8. Lou TY, Wenner HA. Natural and experimental infection of dogs
adenovirus (Toronto A 26/61) with an outbreak of laryngotracheitis with reovirus type 1: pathogenicity of the strain for other animals.
(“kennel cough”): a preliminary report. Can Vet J. 1962;3:238-247. Am J Hygiene. 1963;77:293-304.
2. Renshaw RW, Zylich NC, Laverack MA, et  al. Pneumovirus in 9. Benetka V, Weissenbock H, Kudielka I, et  al. Canine adenovi-
dogs with acute respiratory disease. Emerg Infect Dis. 2010;16:993-995. rus type 2 infection in four puppies with neurological signs. Vet Rec.
3. Edinboro CH, Ward MP, Glickman LT. A placebo-controlled 2006;158:91-94.
trial of two intranasal vaccines to prevent tracheobronchitis (ken- 10. Buonavoglia C, Martella V. Canine respiratory viruses. Vet Res.
nel cough) in dogs entering a humane shelter. Prev Vet Med. 2007;38:355-373.
2004;62:89-99.
180 SECTION 1  Viral Diseases

11. Appel MJ, Menegus M, Parsonson IM, et al. Pathogenesis of canine 36. Percopo CM, Dubovi EJ, Renshaw RW, et al. Canine pneumovirus
herpesvirus in specific-pathogen-free dogs: 5- to 12-week-old pups. replicates in mouse lung tissue and elicits inflammatory pathology.
Am J Vet Res. 1969;30:2067-2073. Virology. 2011;416:26-31.
12. Karpas A, Garcia FG, Calvo F, et  al. Experimental production of 37. Binn LN, Marchwicki RH, Keenan KP, et al. Recovery of reovirus
canine tracheobronchitis (kennel cough) with canine herpesvirus iso- type 2 from an immature dog with respiratory tract disease. Am J
lated from naturally infected dogs. Am J Vet Res. 1968;29:1251-1257. Vet Res. 1977;38:927-929.
13. Ledbetter EC, Dubovi EJ, Kim SG, et  al. Experimental primary 38. Decaro N, Campolo M, Desario C, et al. Virological and molecular
ocular canine herpesvirus-1 infection in adult dogs. Am J Vet Res. characterization of a mammalian orthoreovirus type 3 strain iso-
2009;70:513-521. lated from a dog in Italy. Vet Microbiol. 2005;109:19-27.
14. Ledbetter EC, Kim SG, Dubovi EJ, et al. Experimental reactivation 39. MacLachlan NJ, Dubovi EJ. Coronaviridae. In: Fenner’s Veteri-
of latent canine herpesvirus-1 and induction of recurrent ocular dis- nary Virology. 4th ed. Philadelphia, PA: Elsevier; 2011:393-413.
ease in adult dogs. Vet Microbiol. 2009;138:98-105. 40. Erles K, Toomey C, Brooks HW, et al. Detection of a group 2 coro-
15. Gadsden BJ, Maes RK, Wise AG, et al. Fatal canid herpesvirus 1 navirus in dogs with canine infectious respiratory disease. Virology.
infection in an adult dog. J Vet Diagn Invest. 2012;24:604-607. 2003;310:216-223.
16. Webster RG, Bean WJ, Gorman OT, et al. Evolution and ecology 41. Bryan HM, Darimont CT, Paquet PC, et al. Exposure to infectious
of influenza A viruses. Microbiol Rev. 1992;56:152-179. agents in dogs in remote coastal British Columbia: possible sentinels of
17. Crawford PC, Dubovi EJ, Castleman WL, et  al. Transmission of diseases in wildlife and humans. Can J Vet Res. 2011;75:11-17.
equine influenza virus to dogs. Science. 2005;310:482-485. 42. Knesl O, Allan FJ, Shields S. The seroprevalence of canine respira-
18. Anderson TC, Bromfield CR, Crawford PC, et al. Serological evi- tory coronavirus and canine influenza virus in dogs in New Zea-
dence of H3N8 canine influenza-like virus circulation in USA dogs land. N Z Vet J. 2009;57:295-298.
prior to 2004. Vet J. 2012;191:312-316. 43. An DJ, Jeong W, Yoon SH, et al. Genetic analysis of canine group
19. Barrell EA, Pecoraro HL, Torres-Henderson C, et al. Seroprevalence 2 coronavirus in Korean dogs. Vet Microbiol. 2010;141:46-52.
and risk factors for canine H3N8 influenza virus exposure in house- 44. Zicola A, Jolly S, Mathijs E, et  al. Fatal outbreaks in dogs asso-
hold dogs in Colorado. J Vet Intern Med. 2010;24:1524-1527. ciated with pantropic canine coronavirus in France and Belgium.
20. Crawford C, Spindel M. Canine influenza. In: Miller L, Hurley K, J Small Anim Pract. 2012;53:297-300.
eds. Infectious Disease Management in Animal Shelters. Ames, IA: 45. Buonavoglia C, Decaro N, Martella V, et  al. Canine coronavirus
Wiley-Blackwell; 2009:173-180. highly pathogenic for dogs. Emerg Infect Dis. 2006;12:492-494.
21. Lin Y, Zhao Y, Zeng X, et al. Genetic and pathobiologic charac- 46. Decaro N, Campolo M, Lorusso A, et  al. Experimental infection
terization of H3N2 canine influenza viruses isolated in the Jiangsu of dogs with a novel strain of canine coronavirus causing systemic
Province of China in 2009-2010. Vet Microbiol. 2012;158:247-258. disease and lymphopenia. Vet Microbiol. 2008;128:253-260.
22. Song D, Kang B, Lee C, et al. Transmission of avian influenza virus 47. Ledbetter EC, Hornbuckle WE, Dubovi EJ. Virologic survey of
(H3N2) to dogs. Emerg Infect Dis. 2008;14:741-746. dogs with naturally acquired idiopathic conjunctivitis. J Am Vet
23. Kim H, Song D, Moon H, et al. Inter- and intraspecies transmission Med Assoc. 2009;235:954-959.
of canine influenza virus (H3N2) in dogs, cats, and ferrets. Influ- 48. Ledbetter EC, Kim SG, Dubovi EJ. Outbreak of ocular disease asso-
enza and Other Respiratory Viruses. May 2012. ciated with naturally-acquired canine herpesvirus-1 infection in a
24. Song D, Moon HJ, An DJ, et al. A novel reassortant canine H3N1 closed domestic dog colony. Vet Ophthalmol. 2009;12:242-247.
influenza virus between pandemic H1N1 and canine H3N2 influ- 49. Ledbetter EC, Riis RC, Kern TJ, et al. Corneal ulceration associ-
enza viruses in Korea. J Gen Virol. 2012;93:551-554. ated with naturally occurring canine herpesvirus-1 infection in two
25. Zhan GJ, Ling ZS, Zhu YL, et  al. Genetic characterization of a adult dogs. J Am Vet Med Assoc. 2006;229:376-384.
novel influenza A virus H5N2 isolated from a dog in China. Vet 50. Gervais KJ, Pirie CG, Ledbetter EC, et  al. Acute primary canine
Microbiol. 2012;155:409-416. herpesvirus-1 dendritic ulcerative keratitis in an adult dog. Vet
26. Lin D, Sun S, Du L, et al. Natural and experimental infection of Ophthalmol. 2012;15:133-138.
dogs with pandemic H1N1/2009 influenza virus. J Gen Virol. 51. Erles K, Shiu KB, Brownlie J. Isolation and sequence analysis of
2012;93:119-123. canine respiratory coronavirus. Virus Res. 2007;124:78-87.
27. Dundon WG, De Benedictis P, Viale E, et al. Serologic evidence of 52. Mitchell JA, Brooks H, Shiu KB, et al. Development of a quantita-
pandemic (H1N1) 2009 infection in dogs. Italy. Emerg Infect Dis. tive real-time PCR for the detection of canine respiratory coronavi-
2010;16:2019-2021. rus. J Virol Methods. 2009;155:136-142.
28. Chen Y, Zhong G, Wang G, et al. Dogs are highly susceptible to 53. Anderson TC, Crawford PC, Katz JM, et  al. Diagnostic perfor-
H5N1 avian influenza virus. Virology. 2010;405:15-19. mance of the canine Influenza A Virus subtype H3N8 hemaggluti-
29. Daly JM, Blunden AS, Macrae S, et  al. Transmission of nation inhibition assay. J Vet Diagn Invest. 2012;24:499-508.
equine influenza virus to English foxhounds. Emerg Infect Dis. 54. Castleman WL, Powe JR, Crawford PC, et al. Canine H3N8 influ-
2008;14:461-464. enza virus infection in dogs and mice. Vet Pathol. 2010;47:507-517.
30. Kirkland PD, Finlaison DS, Crispe E, et  al. Influenza virus 55. Rodriguez-Tovar LE, Ramirez-Romero R, Valdez-Nava Y, et  al.
transmission from horses to dogs, Australia. Emerg Infect Dis. Combined distemper-adenoviral pneumonia in a dog. Can Vet J.
2010;16:699-702. 2007;48:632-634.
31. Yamanaka T, Nemoto M, Tsujimura K, et al. Interspecies transmis- 56. Deshpande M, Abdelmagid O, Tubbs A, et al. Experimental repro-
sion of equine influenza virus (H3N8) to dogs by close contact with duction of canine influenza virus H3N8 infection in young puppies.
experimentally infected horses. Vet Microbiol. 2009;139:351-355. Vet Ther. 2009;10:29-39.
32. Chatziandreou N, Stock N, Young D, et al. Relationships and host 57. Australian Government Department of Agriculture, Fisheries, and
range of human, canine, simian and porcine isolates of simian virus Forestry. http://www.daff.gov.au/aqis/cat-dogs/cat4?name=the USA
5 (parainfluenza virus 5). J Gen Virol. 2004;85:3007-3016. (not including Hawaii). Last accessed January 28, 2013.
33. Ellis JA, Krakowka GS. A review of canine parainfluenza virus 58. Jacobs AA, Theelen RP, Jaspers R, et al. Protection of dogs for 13
infection in dogs. J Am Vet Med Assoc. 2012;240:273-284. months against Bordetella bronchiseptica and canine parainflu-
34. Evermann JF, Lincoln JD, McKiernan AJ. Isolation of a paramyxo- enza virus with a modified live vaccine. Vet Rec. 2005;157:19-23.
virus from the cerebrospinal fluid of a dog with posterior paresis. 59. Kontor EJ, Wegrzyn RJ, Goodnow RA. Canine infectious tra-
J Am Vet Med Assoc. 1980;177:1132-1134. cheobronchitis: effects of an intranasal live canine parainfluenza–
35. Hsiung GD. Parainfluenza-5 virus. Infection of man and animal. Bordetella bronchiseptica vaccine on viral shedding and clinical
Prog Med Virol. 1972;14:241-274. tracheobronchitis (kennel cough). Am J Vet Res. 1981;42:1694-1698.
CHAPTER 17  Canine Viral Respiratory Infections 181

60. Chladek DW, Williams JM, Gerber DL, et  al. Canine parainflu- 63. Welborn LV, DeVries JG, Ford R, et al. 2011 AAHA canine vac-
enza–Bordetella bronchiseptica vaccine immunogenicity. Am J Vet cination guidelines. J Am Anim Hosp Assoc. 2011;47:1-42.
Res. 1981;42:266-270. 64. Tyler KL, Barton ES, Ibach ML, et al. Isolation and molecular char-
61. Emery JB, House JA, Bittle JL, et  al. A canine parainflu- acterization of a novel type 3 reovirus from a child with meningitis.
enza viral vaccine: immunogenicity and safety. Am J Vet Res. J Infect Dis. 2004;189:1664-1675.
1976;37:1323-1327.
62. Mouzin DE, Lorenzen MJ, Haworth JD, et  al. Duration of sero-
logic response to five viral antigens in dogs. J Am Vet Med Assoc.
2004;224:55-60.
CHAPTER 18

Infectious Canine Hepatitis


Jane E. Sykes

The strong antigenic relationship between CAV-1 and CAV-2 is


Overview of Infectious Canine Hepatitis clinically important, because vaccines that contain CAV-2 protect
First Described: First described as infectious canine hepatitis against infection with CAV-1 and vice versa. After the introduc-
(ICH) in 1947 in Sweden by Rubarth1; CAV-1 was first iso- tion of CAV-1 vaccines, ICH largely disappeared, but over the past
lated in chick embryos in 19512 decade it has reemerged, with published reports of disease from
Italy, Switzerland, and the United States.5-8 Disease in Europe has
Cause: Canine adenovirus-1 (family Adenoviridae, genus
been associated with puppy trading from kennels in eastern Euro-
Mastadenovirus)
pean countries and possibly spillover of virus circulating in wild-
Affected Hosts: Dogs, coyotes, foxes, wolves, bears, skunks life.9 In Italy, three outbreaks occurred in shelters in southern Italy,
Geographic Distribution: Worldwide and the others involved purebred puppies imported from Hungary
a few days before the onset of clinical signs. Several of the dogs
Mode of Transmission: Direct contact with infected saliva,
were co-infected with other viruses, such as canine distemper virus
feces and urine, and contaminated fomites
(CDV), CPV, or canine enteric coronavirus. Encephalopathy due
Major Clinical Signs in Dogs: Fever, lethargy, inappetence, vom- to CAV-1 infection was described in nine 5-week-old Labrador
iting, hemorrhagic diarrhea, abdominal pain, dehydration, retriever puppies from Arkansas in the United States, all of which
conjunctivitis, petechial hemorrhages, tachypnea, cough, belonged to the same litter.8 The litter was from an unvaccinated
corneal edema (“blue eye”), rarely icterus or neurologic signs. bitch. These case descriptions confirm that CAV-1 continues to
Differential Diagnoses: Other systemic viral diseases such circulate in the dog population and can result in severe disease in
as parvoviral enteritis and canine distemper, enteric viral young dogs when vaccination does not occur, is improperly timed,
and bacterial infections, hepatotoxicosis (e.g., mush- and stress, co-infections, and overcrowded conditions prevail.
rooms), Rocky Mountain spotted fever, gastrointestinal
foreign body, dietary indiscretion, leptospirosis, porto- Clinical Features
systemic shunting with hepatic encephalopathy, dissemi-
Signs and Their Pathogenesis
nated fungal infections (especially systemic candidiasis),
systemic protozoal infections (especially sarcocystosis, CAV-1 is shed in saliva, feces, and urine, and transmission occurs
toxoplasmosis or African trypanosomiasis), hemic neopla- through direct dog-to-dog contact or contact with contaminated
sia (especially lymphoma). fomites such as hands, utensils, and clothing. Ectoparasites such
as fleas and ticks are also potential mechanical vectors.4 Airborne
Human Health Significance: CAV-1 does not infect humans. transmission does not appear to be important. Initial infection
occurs through the nasopharyngeal, conjunctival, or oropha-
ryngeal route, and the virus replicates within the tonsils, after
Etiology and Epidemiology which it spreads to regional lymph nodes and the bloodstream
via lymphatics. Subsequently, infection of hepatocytes and
Infectious canine hepatitis (ICH) is an uncommonly recog- endothelial cells within a variety of tissues occurs, such as the
nized disease of dogs that is caused by canine adenovirus type 1 lungs, liver, kidneys, spleen, and eye with resultant hemorrhage,
(CAV-1), a non-enveloped, icosahedral double-stranded DNA necrosis, and inflammation. The virus replicates in the nucleus
virus that is antigenically related to CAV-2 (see Chapter 17) (Fig- of host cells, where crystalline arrays of virions form. There is
ure 18-1). CAV-1 also causes disease in wolves, coyotes, skunks, severe condensation and margination of nuclear chromatin, with
and bears, as well as encephalitis in foxes, but the diversity of inclusion body formation (Figure 18-2). The virions are released
wildlife hosts is not as great as that for CDV. Ferrets are not by cell lysis, which leads to tissue injury and disseminated intra-
susceptible.3 ICH has also been referred to as Rubarth’s disease, vascular coagulation (DIC). Within the liver, the virus initially
after Carl Sven Rubarth, a veterinarian who first described the infects Kupffer’s cells and subsequently spreads to hepatocytes.
disease in the late 1940s.1 Over the subsequent 10 years, ICH was Clinical signs generally occur after an incubation period of
described worldwide, including the United States, Canada, United 4 to 9 days, although many dogs probably show no signs of ill-
Kingdom, Australia, Japan, Brazil, and throughout Europe. The ness.4 Three overlapping disease syndromes have been described.
virus can survive for months at room temperature, but should be The first is peracute disease with circulatory collapse, coma, and
readily inactivated by disinfectants with activity against canine death after a brief illness that lasts less than 24 to 48 hours.
parvovirus (CPV).4 Disease most commonly occurs in dogs that The second, most commonly described syndrome is acute dis-
are less than 1 year of age, but was reported in adult dogs before ease, which is associated with high morbidity and reported mor-
widespread vaccination for ICH was introduced. tality rates of around 10% to 30%.4 Dogs with acute disease

182
CHAPTER 18  Infectious Canine Hepatitis 183

FIGURE 18-3  Young adult dog with corneal edema from an Italian shelter outbreak
FIGURE 18-1  Structure of canine adenovirus. The virus is a non-enveloped, lcosahedral of CAV-1 infection. (Courtesy Dr. Nicola Decaro, Department of Veterinary Public Health,
virus with fibers (purple) that radiate outwards from the virion. Faculty of Veterinary Medicine of Bari, Italy.)

as seizures, ataxia, circling, apparent blindness, head pressing,


and nystagmus have been reported in association with CAV-1
encephalitis.7,8 The development of neurologic signs may also
represent hepatic encephalopathy, intracranial thrombosis or
hemorrhage, or, as occurred in one outbreak, concurrent infec-
tion with CDV.7
The antibody response appears 7 days after infection and
limits tissue damage. Viral persistence within the renal glom-
eruli, uveal structures of the eye (the iris and ciliary body), and
the cornea can trigger immune complex formation in dogs that
recover from acute illness. This leads to glomerulonephritis with
proteinuria, severe uveitis, and persistent corneal edema in some
surviving dogs. Glomerulonephritis usually occurs about 1 to
2 weeks after the acute signs resolve. Glomerular lesions contain
deposits of viral antigen, IgG, IgM, and C3.11,12 Infection of
FIGURE 18-2  Infection of hepatocytes and endothelial cells with CAV-1 produces the glomerular endothelium is followed by a persistent tubular
characteristic basophilic intranuclear inclusions surrounded by a clear zone that separates infection, development of interstitial nephritis, and viruria, but
them from the marginated chromatin (arrow). H&E stain. (Courtesy Dr. W. Crowell, College chronic renal failure has not been described. Viral shedding in
of Veterinary Medicine, The University of Georgia and Noah’s Archive, College of Veterinary the urine can occur for up to 6 to 9 months after infection. Ante-
Medicine, The University of Georgia. In Zachary JF, McGavin M. Pathologic Basis of Veteri- rior uveitis is associated with massive influx of inflammatory
nary Disease, 5 ed. St. Louis, MO: Mosby; 2012.) cells into the anterior chamber. Occasionally persistent corneal
edema fails to resolve for months and may be associated with
either recover or die within a 2-week period. The third is a more complications such as glaucoma.13 The Afghan hound is report-
chronic form that occurs in dogs with partial immunity, with edly susceptible to this complication.14
death due to hepatic failure weeks (subacute disease) or months In experimental infections with CAV-1, chronic hepatitis
(chronic infection) after initial infection.10 with extensive fibrosis was observed in some dogs that recovered
Acute disease is variably characterized by the presence of from acute illness, with survival for up to 8 months.10 The virus
fever, tonsillitis, conjunctivitis, inappetence, lethargy, weakness, could not be found in hepatic lesions from these dogs. Attempts
polydipsia, vomiting, hematemesis, diarrhea, cough, tachypnea, to detect CAV-1 in the liver of other dogs with chronic active
and icterus. Diarrhea may contain frank blood or melena. Wide- hepatitis using PCR assays have to date been unrewarding.15-17
spread petechial and ecchymotic hemorrhages and hematuria
can be seen. Corneal edema (“blue eye”) occurs in the first week Physical Examination Findings
of illness and results from replication of virus within corneal Physical examination findings in dogs with acute ICH vary, but
endothelial cells (Figure 18-3). Rarely, neurologic signs such might include lethargy, dehydration, fever (up to 106°F or 41°C),
184 SECTION 1  Viral Diseases

TABLE 18-1
Diagnostic Assays Available for Infectious Canine Hepatitis
Assay Specimen Type Target Performance
Histopathology Usually necropsy Hepatic necrosis with in- True sensitivity and specificity of inclusion visualization
­specimens, but also tranuclear inclusions; unknown. Liver biopsy may not be feasible because of
liver biopsy CAV-1 antigen with coagulopathies.
immunohistochemistry
Polymerase chain Blood; rectal, CAV-1 DNA Sensitivity and specificity may vary depending on assay
reaction (PCR) ­conjunctival and design. Specific CAV-1 assays are not widely offered by
nasal swabs; urine; commercial veterinary diagnostic laboratories. Some
tissues collected at assays differentiate between vaccine (CAV-2) and field
necropsy virus (CAV-1). The significance of a positive result from
urine may be difficult to interpret due to subclinical
shedding. False-negative results may occur as a result of
PCR assay inhibition by components of feces or in dogs
with subacute or chronic presentations.
Virus isolation All body secretions, CAV-1 Sensitive and specific, but generally only available as a
tissues research tool.

CAV, Canine adenovirus.

congestion and enlargement of the tonsils (which may be severe), only retrospective diagnosis, and interpretation of these assays
pallor, conjunctivitis, peripheral lymphadenopathy, tachypnea, may be a challenge when there is a history of vaccination.
increased lung sounds, and tachycardia. In some reports, serous
to mucopurulent ocular and nasal discharge have been observed, Laboratory Abnormalities
but these may have resulted from co-infections with other respi- Complete Blood Count
ratory viruses.4 Abdominal palpation may reveal hepatomegaly, Findings reported on the CBC are variable and include leuko-
splenomegaly, or abdominal pain. Icterus is uncommon but can penia, anemia, increased nucleated red blood cells, and mod-
occur in dogs with a more prolonged course of disease. Peripheral erate to severe thrombocytopenia. Leukopenia occurs early in
edema that involves the head, neck, and ventral abdomen has the course of infection and may be profound. Initially there is
been described.18 Puppies with CAV-1 encephalitis may show a lymphopenia, after which neutropenia occurs and worsens
signs of circling, vocalization, head pressing, ataxia, and blind- progressively until death.18 Increased band neutrophils and
ness.8 Coagulopathies may be manifested as cutaneous or muco- toxic changes may also be present. Occasionally leukocytosis is
sal petechial hemorrhages; gingival hemorrhages; epistaxis; or observed.19 Leukocytosis and lymphocytosis may occur as part
prolonged bleeding from venipuncture sites. of the recovery process.
Ocular complications occur in at least 20% of affected dogs.
Unilateral or, less commonly, bilateral corneal edema may be Serum Biochemical Tests
observed, which initially develops at the limbus and is occasion- Changes on the serum biochemistry profile include increased
ally associated with blepharospasm, photophobia, and a serous activity of serum ALT (sometimes >1000 U/L) and ALP, hyper-
ocular discharge.13 Unilateral involvement may progress to bilirubinemia, hypoglycemia, and hypoalbuminemia.20 Determi-
bilateral involvement over several days. Using a slit lamp, the nation of serum ammonia concentration may facilitate diagnosis
cornea is markedly thickened, and there is episcleral and ciliary of hepatic encephalopathy in dogs with neurologic signs.
injection. Uveitis may also be apparent. Corneal ulceration and
increased intraocular pressure has also been described in dogs Urinalysis
with corneal edema, the latter of which may result in blindness.13 The urinalysis of dogs with ICH may reveal proteinuria, hyaline
and granular cylindruria, hematuria, and bilirubinuria.
Diagnosis
Coagulation Profile
ICH should be suspected in any dog less than 1 year of age that In addition to thrombocytopenia, coagulation abnormalities
has a questionable vaccination history and signs of fever, respi- reported in ICH reflect the presence of DIC and hepatic failure
ratory, gastrointestinal, and hepatic disease, and certainly in and include prolonged prothrombin time, markedly prolonged
any young dog that develops corneal edema. Diagnosis is easily activated partial thromboplastin time (to 6 to 7 times control
achieved at necropsy when characteristic intranuclear inclusion values), decreased factor VIII activity, hypofibrinogenemia, and
bodies are seen in tissues, but the sensitivity and specificity of this increased fibrinogen degradation products. Platelet function
finding is unknown. Inclusion bodies may be seen on impression assays have shown reduced platelet adhesion.18
smears of liver biopsies or tissue obtained at necropsy. At the
time of writing, antemortem diagnosis is challenging owing to Diagnostic Imaging
the lack of commercially available assays that specifically detect Findings on plain radiography and abdominal sonography in
the virus or viral DNA, and the disease may be underdiagnosed dogs with ICH have not been reported. Plain radiography might
(Table 18-1). Serologic assays are available, but they provide reveal a normal to slightly enlarged liver, and poor detail as a
CHAPTER 18  Infectious Canine Hepatitis 185

result of abdominal effusion or the young age (and therefore


low intra-abdominal fat content) of affected dogs.

Microbiologic Tests
Virus Isolation
CAV-1 can be readily isolated in a variety of cell types, such
as Madin-Darby canine kidney cells.7,8 In dogs with acute ill-
ness, any body fluid or tissue is likely to contain sufficient virus
for isolation. Cultures are evaluated for a cytopathic effect with
intranuclear inclusions, and the presence of CAV-1 is confirmed
using immunostaining. Isolation is not widely offered by com-
mercial veterinary diagnostic laboratories.

Serologic Diagnosis
Serologic tests are available commercially for detection of IgG
and IgM against CAV-1, which include ELISA assays, hem­
agglutination-inhibition, and serum neutralization. Unfortu-
nately, dogs with acute disease may die before they develop
antibodies to the virus.10 For dogs that recover from illness,
a recent history of vaccination may complicate interpretation
of acute and convalescent phase serology. In the absence of a FIGURE 18-4  Infectious canine hepatitis, hepatic necrosis, liver, dog. The liver from
vaccination history, a fourfold rise in titer over a 2- to 3-week dogs with ICH can be slightly enlarged and friable with a blotchy yellow discoloration.
period together with compatible clinical signs is supportive of Sometimes fibrin is evident on the capsular surface. Note the petechiae on the serosal sur-
face of the intestines caused by vascular damage. (Courtesy Dr. M.D. McGavin, College of
the diagnosis of ICH. Titers that follow natural infection may
Veterinary Medicine, University of Tennessee. In Zachary JF, McGavin MD. Pathologic Basis
be higher than those that follow vaccination. of Veterinary Disease, 5 ed. St. Louis, MO: Mosby; 2012.)
Molecular Diagnosis Using the Polymerase Chain Reaction
Conventional PCR assays for detection of CAV-1 in clinical speci- inflammatory cell infiltrate (see Figure 18-2). Fibrosis may be
mens such as nasal, rectal, and ocular swabs and blood, as well as observed in dogs with chronic liver injury. Interstitial nephri-
tissue obtained at necropsy, have been described. These include tis, with focal accumulations of neutrophils, mononuclear cells,
assays that differentiate between CAV-1 and CAV-221 and repre- and fibrosis, may also be present,22 as well as evidence of wide-
sent one of the most rapid means of antemortem diagnosis. Because spread hemorrhage, thrombosis, and necrosis as a result of DIC.
of the rarity of the disease, the clinical sensitivity and specificity Findings in dogs with CAV-1 encephalitis have included mild
of these assays are not well understood. Real-time PCR assays spongiosis, neuronal necrosis, hemorrhage, and perivascular
that specifically detect CAV-1 infection were not available at the cuffing with mononuclear cells.8 Viral inclusion bodies may be
time of writing (see Chapter 5 for a discussion of the differences found in endothelial cells of meningeal vessels, the cornea, renal
between conventional and real-time PCR assays). The results of glomeruli, and the tonsils. Immunohistochemistry can be used
PCR assays on urine may be more difficult to interpret than those to confirm the presence of the virus within tissues.
for other specimens, because of the potential for chronic shedding
from this site in the absence of clinical signs. Assays that specifi- Treatment and Prognosis
cally detect CAV-1 differentiate between virulent CAV-1 virus and
vaccine virus, because vaccines for ICH all contain only CAV-2. Supportive Care
Treatment of dogs with acute ICH is purely supportive and con-
Pathologic Findings sists primarily of fluid therapy, including crystalloid fluids and
Gross Pathologic Findings blood products. Fluid therapy should be aggressive with careful
Gross pathologic findings in dogs with ICH include blood- monitoring and avoidance of overhydration, because of increased
tinged ascites or hemoabdomen; a slightly enlarged, congested vascular permeability and hypoalbuminemia. Fluids should be
or mottled liver; mild splenomegaly; enlarged, congested, and supplemented with electrolytes and dextrose as required. Other
edematous lymph nodes; and fibrin deposition on the surface of medications that may be indicated include antiemetics, antacids,
abdominal viscera (Figure 18-4). The gallbladder wall is typi- sucralfate, whole blood or plasma transfusions, and colloids such
cally markedly thickened and edematous. Petechial and ecchy- as hetastarch. Partial or total parenteral nutrition may be indicated
motic subserosal hemorrhages may be apparent in multiple for severely affected dogs that do not tolerate enteral feeding.
viscera, and the intestinal tract may contain bloody fluid. The Dogs with DIC may require treatment with heparin in addition
brain may also contain petechial hemorrhages or areas of gray to plasma. Management of hepatic encephalopathy with lactulose
discoloration.8 Parenchymal organs may contain fibrin thrombi. enemas, oral lactulose (in the absence of vomiting), and poorly
Occasionally, multifocal pulmonary consolidation and/or sero- absorbed oral antimicrobial drugs such as ampicillin may also be
sanguineous pleural fluid is noted. indicated. The use of parenteral broad-spectrum antimicrobial
drugs should be considered for dogs with hemorrhagic gastroen-
Histopathologic Findings teritis that may develop bacteremia as a result of bacterial translo-
Histopathologic findings are variable and depend on the course cation. After fluorescein staining has shown no evidence of corneal
of infection and possibly the virus strain. The most characteris- ulceration, dogs with severe corneal edema and uveitis should be
tic finding is hepatocellular necrosis and intranuclear viral inclu- treated with topical ophthalmic preparations that contain gluco-
sion bodies within Kupffer’s cells and hepatocytes, and a mixed corticoids and atropine to prevent development of glaucoma.
186 SECTION 1  Viral Diseases

Prognosis depends on the severity of disease, which reflects 3. Hudson JR, Mansi W. Rubarth’s disease (canine virus hepatitis)
the immune status of the affected dog and possibly the virus II. The insusceptibility of ferrets to experimental infection. J Comp
strain. There is a possibility that recovered dogs may develop Pathol. 1953;63:335-345.
chronic hepatitis or chronic glomerulonephritis, but the extent 4. Cabasso VJ. Infectious canine hepatitis virus. Ann N Y Acad Sci.
1962;101:498-514.
to which this truly occurs is unknown.
5. Pratelli A, Martella V, Elia G, et  al. Severe enteric disease in an
animal shelter associated with dual infections by canine adenovirus
Immunity and Vaccination type 1 and canine coronavirus. J Vet Med B Infect Dis Vet Public
Health. 2001;48:385-392.
Immunity to natural infection with CAV-1 is probably lifelong. 6. Muller C, Sieber-Ruckstuhl N, Decaro N, et al. [Infectious canine
Effective vaccines have been available and widely used as part hepatitis in 4 dogs in Switzerland]. Schweiz Arch Tierheilkd.
of core vaccine programs for dogs for many years. Immunity 2010;152:63-68.
after immunization with attenuated live vaccines lasts at least 7. Decaro N, Campolo M, Elia G, et al. Infectious canine hepatitis: an
3 years and probably longer. Early vaccines, which contained “old” disease reemerging in Italy. Res Vet Sci. 2007;83:269-273.
CAV-1, were associated with the development of corneal edema 8. Caudell D, Confer AW, Fulton RW, et al. Diagnosis of infectious
canine hepatitis virus (CAV-1) infection in puppies with encepha-
and glomerulonephritis in a small percentage (<1%) of immu-
lopathy. J Vet Diagn Invest. 2005;17:58-61.
nized dogs. Replacement of attenuated live CAV-1 with CAV-2 9. Decaro N. Infectious canine hepatitis—a re-emerging disease.
occurred after 1980 and eliminated this complication.23 These Sevilla, Spain: 21st ECVIM-CA Congress; 2011:78–79.
vaccines have the potential to cause transient respiratory signs 10. Gocke DJ, Preisig R, Morris TQ, et al. Experimental viral hepatitis
and tonsillitis in dogs if accidental inhalation occurs, so care in the dog: production of persistent disease in partially immune
should be taken not to aerosolize the vaccine during administra- animals. J Clin Invest. 1967;46:1506-1517.
tion. Maternal antibody persists until puppies are 12 weeks of 11. Morrison WI, Nash AS, Wright NG. Glomerular deposition of
age and interferes with immunization when virus neutralization immune complexes in dogs following natural infection with canine
titers exceed 1:100. Vaccines should be administered every 3 to adenovirus. Vet Rec. 1975;96:522-524.
4 weeks from 6 weeks of age, with the last vaccine given no ear- 12. Hervas J, Gomez-Villamandos JC, Perez J, et al. Focal mesangial-
sclerosing glomerulonephritis and acute-spontaneous infectious
lier than 16 weeks of age (see Appendix). Vaccine virus may be
canine hepatitis: structural, immunohistochemical and subcellular
shed from the respiratory tract by dogs after vaccination, which studies. Vet Immunol Immunopathol. 1997;57:25-32.
has the potential to immunize other dogs as well. It has been 13. Curtis R, Barnett KC. The ocular lesions of infectious canine hepa-
suggested that this phenomenon may have been responsible for titis. 1. Clinical features. J Small Anim Pract. 1973;14:375-389.
the virtual disappearance of the disease in the dog population in 14. Curtis R, Barnett KC. Canine adenovirus-induced ocular lesions in
regions where vaccination is widely performed.24 the Afghan hound. Cornell Vet. 1981;71:85-95.
15. Chouinard L, Martineau D, Forget C, et  al. Use of polymerase
Prevention chain reaction and immunohistochemistry for detection of canine
­adenovirus type 1 in formalin-fixed, paraffin-embedded liver
The best means of prevention of ICH is proper vaccination. of dogs with chronic hepatitis or cirrhosis. J Vet Diagn Invest.
1998;10:320-325.
Additional control measures that could be considered in loca-
16. Boomkens SY, Slump E, Egberink HF, et  al. PCR screening for
tions where outbreaks occur, such as in shelters, include proper candidate etiological agents of canine hepatitis. Vet Microbiol.
disinfection, isolation, and prevention of overcrowding and 2005;108:49-55.
other co-infections, which may worsen disease. Because contact 17. Bexfield NH, Andres-Abdo C, Scase TJ, et  al. Chronic hepatitis
with wild animal species such as coyotes, wolves, and foxes that in the English springer spaniel: clinical presentation, histological
might be shedding the virus may also be a source of infection for description and outcome. Vet Rec. 2011;169(16):415.
dogs, exclusion of these species from interactions with domestic 18. Wigton DH, Kociba GJ, Hoover EA. Infectious canine hepatitis:
dogs may also serve to prevent the disease. animal model for viral-induced disseminated intravascular coagula-
tion. Blood. 1976;47:287-296.
19. Kobayashi Y, Ochiai K, Itakura C. Dual infection with canine dis-
Public Health Aspects temper virus and infectious canine hepatitis virus (canine adenovirus
type 1) in a dog. J Vet Med Sci. 1993;55:699-701.
There is no evidence that CAV-1 infects humans. 20. Beckett SD, Burns MJ, Clark CH. A study of the blood glucose,
serum transaminase, and electrophoretic patterns of dogs with
SUGGESTED READINGS infectious canine hepatitis. Am J Vet Res. 1964;25:1186-1190.
Decaro N, Campolo M, Elia G, et  al. Infectious canine hepatitis: an 21. Hu RL, Huang G, Qiu W, et al. Detection and differentiation of
“old” disease reemerging in Italy. Res Vet Sci. 2007;83:269-273. CAV-1 and CAV-2 by polymerase chain reaction. Vet Res Com-
Gocke DJ, Preisig R, Morris TQ, et al. Experimental viral hepatitis in mun. 2001;25:77-84.
the dog: production of persistent disease in partially immune animals. 22. Wright NG. Interstitial nephritis in a dog associated with infectious
J Clin Invest. 1967;46:1506-1517. canine hepatitis virus. Vet Rec. 1970;86:92-93.
23. Bass EP, Gill MA, Beckenhauer WH. Evaluation of a canine adeno-
REFERENCES virus type 2 strain as a replacement for infectious canine hepatitis
1. Rubarth S. An acute virus disease with liver lesion in dogs (hepatitis vaccine. J Am Vet Med Assoc. 1980;177:234-242.
contagiosa canis): a pathologico-anatomical and etiological investi- 24. MacLachlan NJ, Dubovi EJ. Adenoviridae. Fenner’s Veterinary
gation. Acta Path Microbiol Scand. 1947(Suppl):69. Virology. 4th ed. Elsevier; 2011:203-212.
2. Miles JA, Parry HB, Larin NM, et al. Cultivation of canine hepati-
tis virus in embryonated hen’s eggs and its subsequent transmission
to dogs. Nature. 1951;168(4277):699-700.
CHAPTER 19
Feline Panleukopenia Virus Infection
and Other Viral Enteritides
Jane E. Sykes

have been reported in households of fully vaccinated kittens,


Overview of Feline Parvoviral Enteritis possibly because of exposure to large amounts of virus in the
First Described: 1928, Verge and Christoforoni, France1 environment.4 Outbreaks of panleukopenia in cats correlate
seasonally with increases in susceptible newborn kitten num-
Cause: Feline panleukopenia virus (FPV); also canine parvo-
bers. Panleukopenia occurs most commonly in multicat house-
virus (CPV)-2a, CPV-2b, and CPV-2c (Family Parvoviridae,
holds, and especially in enclosed, shelter environments. It can
subfamily Parvovirinae, genus Parvovirus)
also occur in cats with outdoor exposure, such as barn, feral,
Affected Hosts: Domestic and wild cats; foxes, mink, and and stray cats. In one study, the prevalence of protective anti-
raccoons body titers to FPV in feral cats in Florida was only 33%, which
Geographic Distribution: Worldwide suggested a low rate of exposure to the virus.5 In some North
American shelters, devastating outbreaks of panleukopenia have
Mode of Transmission: Direct contact with virus in feces and
led to euthanasia of large numbers of cats. Contact with other
vomitus and contaminated fomites.
cats may not be present in the history, because fomite transmis-
Major Clinical Signs: Fever, lethargy, inappetence, vomiting, sion is so effective.3 FPV replicates to a limited extent in dogs,
diarrhea, dehydration, sudden death. Neurologic signs without disease or virus shedding. Some feline panleukopenia
(primarily cerebellar signs) may also occur. results from infection of cats by the related mink enteritis virus,
Differential Diagnoses: Other feline viral enteritides, toxins, CPV-2a, CPV-2b, or CPV-2c.6-8 Mixed infections with FPV and
gastrointestinal foreign body, enteric parasitic infections CPV-2 variants have been detected in cats, and there is evidence
such as giardiasis and nematode infections, enteric bac- for recombination between FPV and CPV-2 variants.7,9 Infec-
terial infections such as salmonellosis, pancreatitis, or tion of cats with CPV-2 variants is uncommon in Europe but
inflammatory bowel disease. Congenital central nervous predominated among cats with panleukopenia in Asia.10
system defects should be considered in cats with neuro- Other viral pathogens that have been associated with gas-
logic signs. troenteritis in cats include feline enteric coronavirus (see
Chapter 20), FeLV, rotaviruses, caliciviruses, reoviruses, and
Human Health Significance: FPV is not known to infect astroviruses. A torovirus-like agent has been associated with
humans, but was isolated from a monkey. a ­syndrome of diarrhea and protrusion of the nictating mem-
branes in cats.11 Togavirus-like and picornavirus-like particles
have been identified in the feces of Australian cats, but their
Etiology and Epidemiology significance is uncertain.12

Feline panleukopenia virus (FPV) is a parvovirus that causes Clinical Features


enteritis and panleukopenia in domestic and wild cat species
worldwide.2 It has also been associated with disease in rac- Signs and Their Pathogenesis
coons, mink, foxes, and a monkey, and can replicate in ferrets The pathogenesis of FPV infection is similar to that of CPV
without causing disease. Feline panleukopenia is sometimes infection (see Chapter 14). Transmission is by the fecal-oral
confusingly referred to as “cat plague” and “feline distemper.” route, and indirect transmission through contaminated fomites
FPV is a small, single-stranded non-enveloped DNA virus that represents the most important means of infection. Like CPV,
is closely related to CPV-2, but in contrast to CPV-2, which FPV enters cells using transferrin receptors13 and replicates in
emerged in the late 1970s, the existence of FPV has been known cells that are in the S-phase of the mitotic cycle. Initially, the
since the 1920s.1 FPV has the same ability as CPV-2 to survive virus replicates in oropharyngeal lymphoid tissue, after which
long periods in the environment and resist disinfection, and has it disseminates in blood to all tissues. Infection of lymphoid tis-
the same preference for replication in rapidly dividing cells (see sues leads to lymphoid tissue necrosis. Infection of the marrow
Chapter 14). is associated with leukopenia, which is compounded by neutro-
Feline panleukopenia is most likely to occur in cats younger phil sequestration in damaged gastrointestinal tissue. The virus
than 1 year of age, but it can occur in unvaccinated or improp- replicates in the intestinal crypt epithelial cells, with shortening
erly vaccinated cats of all ages. The median age of affected cats of villi, increased intestinal permeability, and malabsorption.
in one study was 4 months, and when disease occurred in vac- Subclinical infection is probably widespread, especially in
cinated cats, it occurred only in cats that had not received a young adult or adult, immune-competent cats. Disease severity
booster vaccine after 12 weeks of age.3 However, kitten deaths depends on factors such as age, immune status, and concurrent

187
188 SECTION 1  Viral Diseases

infections with other bacterial or viral pathogens, which increase Kittens with cerebellar signs are generally bright and alert
the turnover rate of intestinal epithelial cells and enhance viral but exhibit intention tremors, incoordination, ataxia, hyper-
replication and cellular destruction. Co-infections can occur with metria, a broad-based stance, decreased postural reactions, a
feline enteric coronavirus, Clostridium piliforme, Salmonella truncal sway, and absence of a menace response. Cats with fore-
spp., FeLV, or astroviruses.14-17 Disease generally occurs after an brain disease may show abnormal behavior, such as aggression
incubation period of 2 to 10 days. The peracute form of disease or decreased mentation. Examination of the ocular fundus may
involves death without apparent premonitory signs. Infection of reveal folding of the retina, evidence of retinal degeneration
kittens or adult cats results in clinical signs of fever, lethargy, with discrete gray spots, and optic nerve hypoplasia. Retinal
vocalization, weakness, and inappetence, which may progress lesions may be an incidental finding in older, recovered cats,
to profound dehydration, vomiting, sometimes watery to hem- which includes surviving cats with cerebellar hypoplasia.
orrhagic diarrhea, and rapid loss of weight. Some cats develop
only anorexia and lethargy, in the absence of vomiting, diar- Diagnosis
rhea, or leukopenia.3 Secondary bacterial infections appear to be
essential for signs of disease to occur. Death ­usually results from Laboratory Abnormalities
complications relating to dehydration, electrolyte imbalances, Complete Blood Count
hypoglycemia, hemorrhage, or bacteremia and endotoxemia. The most common abnormality on the CBC in feline panleu-
In the developing fetus or neonate, FPV replicates in a vari- kopenia is leukopenia, which is due to a neutropenia and lym-
ety of tissues. Abortion, congenital abnormalities, or infertil- phopenia (Box 19-1).3 Total leukocyte counts may be as low
ity can result from infection early in pregnancy, although the as 50 cells/µL, and toxic band neutrophils may be present. In
queen is generally otherwise unaffected. Later in pregnancy or one study, only 65% of 187 cats with panleukopenia were leu-
in neonates up to approximately 1 week of age, viral destruc- kopenic, so an absence of leukopenia does not rule out FPV
tion of Purkinje cells and granule precursor cells located in the infection. Recovery may be associated with lymphocytosis and
cerebellar external granular layer leads to cerebellar hypoplasia
(Figure 19-1). The severity of infection can vary between kittens
in a litter. Sometimes a portion of the litter fails to survive and
the remainder develop neurologic signs.18 Signs of cerebellar
ataxia are nonprogressive and are most apparent when kittens
begin to walk at about 2 to 3 weeks of age, although menta-
tion and appetite are otherwise normal, and these kittens can
sometimes make acceptable pets. Other developmental central
nervous system (CNS) abnormalities have been reported less
commonly, and include hydrocephalus, porencephaly (cystic
lesions within the cerebral hemispheres), or hydranencephaly
(complete replacement of the cerebral hemispheres with cystic
lesions). These abnormalities may be accompanied by signs of
forebrain damage, such as seizures and behavioral changes.
Unlike canine parvoviruses, FPV appears to be able to infect
neurons other than the cerebellar Purkinje cells, which are ter-
minally differentiated cells.19 Ocular lesions can also develop
and include retinal folding, dysplasia and degeneration, and A
optic nerve hypoplasia.
The DNA of FPV has been detected in the myocardium of
cats with hypertrophic, dilated, and restrictive cardiomyopathy,
but was not detected in a subset of healthy control cats.20 Addi-
tional studies to assess the role of FPV in feline myocarditis and
cardiomyopathy are needed.
Fecal shedding of virus usually lasts for several days, and in
some cats, it may persist for up to 6 weeks. Kittens infected in
utero have been reported to develop immune tolerance to the
virus, with viral persistence in the kidneys and lungs for up to
1 year in the absence of shedding.

Physical Examination Findings


The most common physical examination findings are weakness,
lethargy, and dehydration. Fever (103°F to 107°F or 39.5°C to
42.5°C) may be present early in the course of illness. Pain may
be noted on palpation of the abdomen, or a hunched posture B
may be present. The perianal region may be contaminated with FIGURE 19-1  A, Severe cerebellar hypoplasia in a 1-year-old male neutered domes-
feces. Oral ulceration and mucosal pallor may be present in tic shorthair that was euthanized for cerebellar signs and seizures. The neurologic signs
severely affected cats, and rarely, bacteremia may be accompa- had been present since the cat was found at approximately 4 months of age. There is a
nied by icterus. Terminally, affected cats may be hypothermic, paucity of neurons and glial cells in all layers. B, Normal feline cerebellum. Note the dra-
bradycardic, and comatose. matically increased width of the molecular layer when compared to (A).
CHAPTER 19  Feline Panleukopenia Virus Infection and Other Viral Enteritides 189

leukocytosis. Thrombocytopenia and mild anemia are also com- Serum Biochemical Tests
mon. Thrombocytopenia may result from damage to the mar- Serum biochemistry analysis may show hypoalbuminemia,
row or, possibly, disseminated intravascular coagulation (DIC). hypoglobulinemia, and/or hypocholesterolemia; electrolyte
abnormalities such as hyponatremia or hypernatremia, hypo-
chloremia, hyperkalemia, or, less commonly hypokalemia; and
BOX 19-1 acid-base abnormalities (see Box 19-1). In severely affected cats,
azotemia, increased serum activities of AST or ALT, or hyper-
Prevalence of Laboratory Abnormalities in Cats with bilirubinemia may be present. Hyperglycemia or hypoglycemia
Panleukopenia may also be identified.

Diagnostic Imaging
Leukopenia: 122/187 (65%) Plain Radiography
Thrombocytopenia: 83/153 (54%) As in dogs with parvoviral enteritis, abdominal radiography
Anemia: 91/187 (48%) in cats with panleukopenia may show evidence of poor serosal
Neutropenia: 64/137 (47%) detail and a fluid- and gas-filled gastrointestinal tract.
Lymphopenia: 53/137 (39%)
Hypoalbuminemia: 45/101 (45%) MRI Findings
Hypochloremia: 40/112 (36%) MRI of cats with neurologic signs due to FPV may reveal evi-
Hyponatremia: 41/127 (32%) dence of cerebellar agenesis or hypoplasia. Rarely, hydrocepha-
Hypoproteinemia: 46/153 (30%) lus, porencephaly, or hydranencephaly can be detected.18
Hyperglycemia: 48/168 (29%)
Increased AST activity: 26/98 (27%) Microbiologic Tests
Hyperkalemia: 30/132 (23%) Diagnostic assays available for feline panleukopenia are listed
Increased BUN: 36/168 (21%) in Table 19-1.
Hyperbilirubinemia: 19/134 (14%)
Increased ALT activity: 18/135 (13%) Serologic Diagnosis
Increased creatinine: 12/155 (8%) Use of serology for diagnosis of feline panleukopenia is com-
Hypokalemia: 9/132 (7%) plicated by widespread exposure or immunization, so serologic
Hypernatremia: 8/127 (6%) assays that detect antibody against FPV are generally used to
Hypoglycemia: 10/168 (6%) assess the need for vaccination rather than for diagnosis. They
can also be used in outbreak situations in order to determine
Modified from Kruse BD, Unterer S, Horlacher K, et al. Prognos- which cats are at risk for development of disease and virus shed-
tic factors in cats with feline panleukopenia. J Vet Intern Med ding, and which cats are protected and therefore at low risk.
2010;24:1271-1276. The gold standard method for FPV serology is hemagglutination

TABLE 19-1
Diagnostic Assays Available for Feline Panleukopenia
Assay Specimen Type Target Performance
Canine parvovirus Feces Parvoviral antigen Sensitivity varies with the assay used and the timing
fecal antigen of specimen collection. False negatives are common,
ELISA but a positive result generally indicates infection.
Histopathology Usually necropsy Crypt necrosis with intra- Can be used for necropsy diagnosis.
specimens, especially nuclear inclusions; FPV
gastrointestinal tissues antigen with IHC or IFA
Polymerase chain Feces, tissue samples FPV DNA Sensitivity and specificity varies depending on assay
reaction (PCR) design. The extent to which attenuated live vaccine
virus can be detected after vaccination is not well
understood. Because of the high sensitivity of some
assays, the significance of a positive result may be dif-
ficult to interpret. False-negative results may occur as
a result of inhibition of PCR by components of feces.
Fecal electron Feces Virus particles Not widely available, turnaround time can be slow,
microscopy and may be expensive. Requires the presence of
large amounts of virus.
Virus isolation Feces, tissues FPV Difficult, not widely available. Used primarily as a
research tool.

FPV, feline panleukopenia virus; IFA, immunofluorescent antibody; IHC, immunohistochemistry.


190 SECTION 1  Viral Diseases

inhibition, which measures the ability of serum to prevent agglu- Molecular Diagnosis Using the Polymerase Chain Reaction
tination of erythrocytes by the virus (see Chapter 2). Serum neu- Specific real-time PCR assays have been developed for detection
tralization assays may also be used. Point-of-care assays designed of FPV and differentiation of FPV from CPV-2 variants and are
to detect antibody titers to CPV have a low sensitivity (28%) offered by veterinary diagnostic laboratories. Clinicians should
for detection of antibodies to FPV in cats.21 A point-of-care contact their laboratory to determine the specificity of the assay
assay designed to detect feline antibodies (ImmunoComb Feline offered.25 These assays can be used on whole blood or feces.
VacciCheck Test Kit, Biogal, Galed Labs, Israel) also had a low The extent to which these assays detect attenuated live vaccine
sensitivity (49%), although specificity was high.21 The use of virus after vaccination requires further study. Assays have also
this test for risk analysis in shelter situations may lead to inap- been developed that differentiate between field and vaccine
propriate removal or isolation of cats with false-negative results strains of FPV.26
for protective antibody titers, which might waste time, space,
and financial resources.21 However, positive results should Pathologic Findings
­reliably indicate protection. The test can be performed quickly Gross Pathologic Findings
(30 minutes) and requires as little as 5 µL of serum or plasma, Gross pathologic findings in feline panleukopenia include thymic
so provided it is understood that a negative result means either involution; thickening, distention, and discoloration of the intes-
a protected or susceptible status, and a positive result equates tinal wall with serosal hemorrhage (Figure 19-2); and enlarged,
to protection, the test still has the potential to provide useful edematous mesenteric lymph nodes. The intestine may contain
information. bloody liquid contents, and mucosal hemorrhage may be identi-
fied. Hemorrhages may be visible on the surface of other organs
Antigen Detection Enzyme Linked Immunosorbent Assay as well. In some cats, mild pleural or peritoneal effusion is pres-
FPV can be detected in feces or rectal swabs using antigen ent. Cats infected prenatally may have cerebellar aplasia, or more
assays designed to detect CPV.22,23 The sensitivity and speci- commonly a small cerebellum (often half to three-quarters normal
ficity of these assays varies from one assay to another and with size).27 Rarely other developmental CNS abnormalities such as
the stage of infection, because virus shedding may be tran- hydrocephalus, hydranencephaly, or porencephaly are observed.18
sient. In general, false-negative results are common with these
assays, but false positives are uncommon, so a positive test Histopathologic Findings
result in a cat with consistent clinical signs suggests a diag- Histopathologic findings in the intestinal tract are similar to those
nosis of feline panleukopenia. The specificity of one point-of- described for CPV-2 infection, with crypt dilation and necrosis
care device (SNAP Parvo, IDEXX Laboratories, Westbrook, of crypt epithelial cells, accumulation of cellular debris, neutro-
ME) was high; 54 of 55 positive assay results were confirmed phil infiltration, loss of villi, and submucosal edema throughout
with a PCR assay. Another study of 52 cats with diarrhea the small and large intestines; the jejunum and ileum are usually
and 148 healthy cats showed variability in the sensitivity most severely affected (Figure 19-3, A). Acutely affected cats
and specificity of five different test kits when compared with show widespread lymphoid depletion and there may be hyper-
fecal electron microscopy.23 Sensitivity ranged from 50% to plasia of mononuclear phagocytes. Intranuclear inclusions are
80%, and specificity ranged from 94% to 100%. This study found in some cats (Figure 19-3, B). Examination of the bone
included only 10 cats with FPV as determined with electron marrow may reveal bone marrow hypoplasia. Examination of
microscopy. Additional studies are warranted that evaluate the cerebellum shows cellular depletion; reactive astrocytosis
the sensitivity and specificity of these assays in larger numbers may be present. Immunohistochemistry or immunofluorescent
of cats with FPV infection when both real-time PCR and elec-
tron microscopy are used as the gold standard. False-positive
fecal antigen assay results after vaccination with attenuated
live viral vaccines appear to be uncommon, but again vary
with the test used.24 Of the SNAP Parvo (IDEXX Laborato-
ries), AGEN CPV (AGEN Biomedical Ltd., Brisbane, Austra-
lia), and the Witness CPV (Synbiotics Corp, San Diego, CA),
the SNAP Parvo was least likely to yield positive results after
vaccination.

Fecal Electron Microscopy


Fecal electron microscopy is still offered by some institutions
for diagnosis of viral enteritis. It may also facilitate diagnosis
of other infections such as rotavirus, astrovirus, torovirus, and
coronavirus infections. Turnaround time may be slow. Gener-
ally speaking, large amounts of virus must be present for results
to be positive, and technical expertise is required to accurately
identify virus in the stool.

Virus Isolation
FPV can be isolated in feline cells, but as with CPV, isolation
can difficult, and the virus shows minimal cytopathic effects. FIGURE 19-2  Intestinal tract of a 2-year-old intact female domestic longhair cat
As a result, isolation of FPV is a specialized procedure that is with severe feline panleukopenia. The intestinal loops are dilated and flaccid and discol-
uncommonly used for diagnosis. ored red to purple. Ruler = 1 cm.
CHAPTER 19  Feline Panleukopenia Virus Infection and Other Viral Enteritides 191

antibody may be used to document the presence of the virus vaccines. An intranasal FPV, FHV-1 and feline calicivirus vac-
within tissues (see Figure 19-3, C). cine is available; its use has been controversial, because panleu-
kopenia is a systemic disease. An outbreak of salmonellosis and
Treatment and Prognosis panleukopenia occurred in one cattery that used an intranasal
FPV vaccine.4 No difference was noted in seroconversion rates
Antimicrobial Treatment and Supportive Care between five cats vaccinated with the intranasal vaccine and five
Treatment of cats with feline panleukopenia is with supportive cats vaccinated with a parenteral attenuated live vaccine, but
care, especially intravenous crystalloids and parenteral antimi- the number of cats in this study was small, and a larger number
crobial drug treatment as for CPV-2 infections (see Chapter 14). of cats that were vaccinated with the parenteral vaccine had
Dextrose supplementation of the fluids may be required, and protective antibody titers on day 7.30
blood glucose concentration should be monitored. Oral intake Both inactivated and attenuated live vaccine types induce
of food and water should be withheld until vomiting has ceased. protective antibody titers after vaccination in a high propor-
Experience with early enteral nutrition has not been reported tion of cats,5 although the attenuated live vaccine may be
in cats, and extreme care is warranted to prevent aspiration more likely to induce protective titers than the inactivated vac-
pneumonia. Antiemetics such as metoclopramide or ondanse- cine.24 Provided maternally-derived antibody (MDA) is absent,
tron may be effective. In contrast to canine parvoviral enteritis, immunity occurs within 1 week after a single vaccination with
treatment with rfIFN-ω has not been beneficial for treatment of an attenuated live viral vaccine31 and lasts for at least 3 years,
feline panleukopenia, although increased antibody production and possibly for life.32 Nevertheless, two doses, 3 to 4 weeks
and a reduced acute inflammatory response was observed.28 apart, have been recommended for initial vaccination with
attenuated live vaccines in the absence of MDA. Two injec-
Prognosis tions are always required for inactivated vaccines, and maxi-
Cats with panleukopenia that survive the first 5 days of treat- mal immunity does not occur until 1 week after the second
ment usually recover, although recovery is often more pro- dose. However, even with an inactivated vaccine, challenge
longed than it is for dogs with parvoviral enteritis. In 244 cats 7.5 years after vaccination was associated with protection.33 The
with feline panleukopenia from Europe, the survival rate was use of inactivated vaccines should be reserved for immunosup-
51.1%.3 Nonsurvivors had lower leukocyte and platelet counts pressed cats or colostrum-deprived neonates that are less than
than survivors, and cats with white cell counts below 1000/µL 4 weeks of age, or when there is a need to vaccinate pregnant cats.
were almost twice as likely to die than those with white cell In shelter situations, the use of attenuated live vaccines is always
counts above 2500/µL. Only total leukopenia, and not lym- recommended because of the slow onset of immunity with inac-
phopenia, was correlated with mortality. Hypoalbuminemia tivated vaccines.34 FPV vaccines protect cats against challenge
and hypokalemia were also associated with an increased risk of with CPV-2b strains.35,36 However, cross-reactivity of antibod-
mortality. In contrast to dogs with parvoviral enteritis, mortal- ies induced by FPV vaccination to CPV-2 strains was lower than
ity in cats does not appear to be correlated with age. that to FPV as determined using hemagglutination inhibition.37
Cerebellar signs in kittens with cerebellar hypoplasia typi- The extent to which FPV vaccines protect against infection with
cally do not progress and may improve slightly as a result of other CPV-2 variants requires further investigation.38
compensatory responses from other senses such as vision.29 The most common reason for vaccine failure is interference
by MDA. Maternal antibody persists until at least 12 weeks,
Immunity and Vaccination and possibly longer in some cats. Virus neutralization titers
above 1:10 are likely to interfere with vaccination, and kittens
Recovery from feline panleukopenia is thought to confer life- with titers below 1:40 are generally considered to be suscep-
long immunity. Effective vaccines are widely available, and tible to infection by FPV. Kittens should be vaccinated every
include both parenteral inactivated and attenuated live viral 3 to 4 weeks from 6 to 8 weeks of age, and it is recommended

A B C
FIGURE 19-3  A, Histopathology of the jejunum from a kitten with panleukopenia. Villi are rounded and blunted with nearly complete epithelial loss and crypt dilation. H&E stain.
B, Histopathology of the jejunum from a young barn cat that died after several days of profound weakness and neurologic signs. Six other cats in the barn died with the same signs. Mucosal
crypts are dilated and contain debris, and intranuclear inclusions are present (small arrows). Another cell (large arrow) is foamy and degenerate and has a hyperchromatic nucleus. H&E
stain. C, Same cat as in B. The presence of FPV in the intestinal tract is confirmed with immunohistochemistry (brown stain). (Courtesy Dr. Patricia Pesavento, University of California, Davis
Veterinary Anatomic Pathology Service.)
192 SECTION 1  Viral Diseases

that the last vaccine in the kitten series be given no earlier 2 mL of type-matched serum from cats with a high antibody
than 14 to 16 weeks of age. When there is a history of an out- titer.39,40 However, this is only effective when administered
break situation, the final booster could be given no earlier than before the onset of clinical signs, and it can interfere with subse-
18 to 20 weeks of age.39 In all situations, a booster should be quent vaccination. For these kittens, it has been recommended
administered at 1 year, and every 3 years thereafter. that vaccination be withheld for 3 weeks after the serum has
Vaccination of pregnant queens with attenuated live viral been administered.39 Passive immunization may be useful when
vaccines can cause cerebellar hypoplasia or fetal losses. The fre- cats are introduced into a shelter situation where a known prob-
quency with which this occurs is unknown. As a result, it has lem exists. Repeated treatment with serum should be avoided
been suggested that pregnant queens only be vaccinated with because hypersensitivity reactions may occur. Prevention of
attenuated live FPV vaccines if they are being introduced into feline panleukopenia should also include proper disinfection
a shelter and quarantine while immunization with inactivated with disinfectants that are effective against parvoviruses, such
vaccines is performed is not possible. Alternatively, assessment as bleach, accelerated hydrogen peroxide, or potassium peroxy-
for protective antibody titers with an in-house test kit (where monosulfate (see Chapter 11) and, in shelter situations, isola-
available) could be performed. tion or removal of cats that develop gastrointestinal illness, and
separate housing for healthy kittens.
Prevention
Public Health Aspects
New kittens should not be introduced into households that
previously contained cats infected with FPV unless they are Although FPV is not known to infect humans, a unique strain of
fully vaccinated. In the face of an outbreak, exposed and sus- FPV was recently isolated from a diarrheic monkey in China.41
ceptible kittens may be effectively protected for 2 to 4 weeks This strain was shown to cause panleukopenia in inoculated
through subcutaneous or intraperitoneal administration of cats.

CASE EXAMPLE Laboratory Findings:


CBC:
HCT 46% (30-50%)
Signalment: “Callie”, a 2 year-old, female intact domestic MCV 49.8 fL (42-53 fL)
longhair from Woodland, CA MCHC 30.4 g/dL (30-33.5 g/dL)
History: Callie was brought to an emergency clinic for acute WBC 150 cells/µL (4500-14,000 cells/µL)
onset of collapse and severe illness. The current owner had Neutrophils 0 cells/µL (2000-9000 cells/µL)
fostered the cat for 1 week after she was found as a stray. The Lymphocytes 141 cells/µL (1000-7000 cells/µL)
cat had been nursing a litter of kittens. The kittens were 4 Highly reactive lymphocytes 6 cells/µL
weeks old, being weaned and apparently healthy. Since be- Monocytes 3 cells/µL (50-600 cells/µL)
ing fostered, the cat had exhibited a progressive decrease Platelets 32,000 platelets/µL (180,000-500,000 platelets/µL).
in appetite and thirst, and her feces had become soft and Serum Chemistry Profile:
pasty. The night before she was brought to the emergency Sodium 141 mmol/L (151-158 mmol/L)
clinic, she had been bathed, and afterwards she vomited Potassium 4.0 mmol/L (3.6-4.9 mmol/L)
bile-stained fluid twice and was placed on a heating pad. Chloride 111 mmol/L (117-126 mmol/L)
The following morning she was found laterally recumbent Bicarbonate 23 mmol/L (15-21 mmol/L)
and minimally responsive. Phosphorus 6.0 mg/dL (3.2-6.3 mg/dL)
Physical Examination: Calcium 7.4 mg/dL (9.0-10.9 mg/dl)
Body Weight: 2.3 kg BUN 24 mg/dL (18-33 mg/dL)
General: Stuporous mentation, estimated to be 8% to 10% Creatinine 0.5 mg/dL (1.1-2.2 mg/dL)
dehydrated, T < 92°F (<33°C), HR = 132 beats/min, RR = 32 Glucose 68 mg/dL (63-118 mg/dL)
breaths/min, mucous membranes pale and tacky, unable Total protein 3.2 g/dL (6.6-8.4 g/dL)
to assess CRT. Fecal and urinary stains were present Albumin 1.6 g/dL (2.2-4.6 g/dL)
around the perineum and on the caudal aspect of the Globulin 1.6 g/dL (2.8-5.4 g/dL)
pelvic limbs. ALT 125 U/L (27-101 U/L)
Musculoskeletal: Body condition score 2/9. Diffuse muscle AST 143 U/L (17-58 U/L)
wasting was present. The cat was laterally recumbent and ALP 4 U/L (14-71 U/L)
nonambulatory. Creatine kinase 2409 U/L (73-260 U/L)
Cardiovascular: Weak femoral pulses. No murmurs or Gamma GT <3 U/L (0-4 U/L)
arrhythmias were detected. Cholesterol 68 mg/dL (89-258 mg/dL)
Gastrointestinal and Urogenital: The abdomen was soft Total bilirubin 0.2 mg/dL (0-0.2 mg/dL)
and nonpainful on palpation. Fluid-filled intestinal loops Magnesium 2.5 mg/dL (1.5-2.5 mg/dL).
were palpated, and the urinary bladder was small (<5 cm) and Imaging: An abdominal ultrasound showed marked fluid dis-
soft. tention of the small intestines.
All Other Systems: No abnormalities were detected. Microbiologic Testing: In-clinic ELISA serology for FeLV anti-
gen and FIV antibody: negative.
CHAPTER 19  Feline Panleukopenia Virus Infection and Other Viral Enteritides 193

Treatment and Outcome: Callie was treated with active to purple and contained a small amount of dark red mucoid
warming, and a central venous access line was placed. Intra- material. The mesenteric lymph nodes were prominent, and
venous crystalloids (four warmed 60-mL boluses of lactated their cut surface was discolored red to dark pink. The liver
Ringer’s solution [LRS], each given over 20 minutes) were ad- was pale yellow and extended beyond the costal arches. Nu-
ministered, after which an venous acid-base panel showed merous petechial hemorrhages were present on the serosal
a pH of 7.267 (7.31-7.46), bicarbonate of 17.5 mmol/L (14- surface of the urinary bladder.
22 mmol/L), base excess of −8.1 mmol/L (−4 to +2 mmol/L), Histopathologic Findings: Within the duodenum, jejunum,
pCO2 of 39.7 mm Hg (25-37 mmHg), lactate of 4.5 mEq/L and ileum there was severe, subacute, diffuse necrotizing
(<2 mEq/L), glucose of 52 mg/dL, potassium of 3.1 mEq/L, and fibrinohemorrhagic enteritis. The villi were necrotic,
sodium of 141 mEq/L, and ionized calcium of 1.17 (1.1-1.4 fused, and markedly blunted with a minimal inflammatory
mmol/L). A dextrose bolus and ticarcillin-clavulanic acid response. Necrosis extended into the crypts, and there was
(22 mg/kg, q6h, IV) were administered. The cat repeatedly some evidence of a regenerative response that included en-
vomited blood-tinged fluid, and so treatment with meto- terocyte hypertrophy, karyomegaly, and rare mitotic figures.
clopramide (0.02 mg/kg/hr) and famotidine (0.5 mg/kg IV Mixed bacteria, which included small gram-negative rods
q12h) was initiated. Aggressive crystalloid fluid therapy was and large numbers of gram-positive cocci, lined and effaced
continued (LRS supplemented with 30 mEq/L KCl and 2.5% the denuded villi. There was diffuse lymphoid depletion in
dextrose), and treatment with hetastarch was also initiated. lymph nodes, as well as the mucosa-associated lymphoid
Systolic blood pressure (Doppler) was 60 to 90 mm Hg, heart tissue in the cecum and colon. Marked depletion of ery-
rate increased to 170 beats/min, and rectal temperature in- throid and myeloid precursors was present in the bone mar-
creased to 100°F. When the CBC results were available, the row. There were multifocal areas of hemorrhage in the skel-
cat was placed in the isolation ward. etal and cardiac muscle, intestinal tract, and lungs, as well
Pasty diarrhea that contained sloughed mucosa occurred as fibrin thrombi in the lungs. FPV antigen was identified in
every 1 to 2 hours, which transitioned to liquid red feces intestinal epithelial cells using immunohistochemistry. Cul-
over 24 hours. A CBC again showed absolute neutropenia ture of the jejunum for Salmonella spp. was negative.
and 15,000 platelets/µL. After another 24 hours, the cat’s Diagnosis: Feline panleukopenia
condition deteriorated despite aggressive treatment and Comments: The cat in this report was a stray with an un-
monitoring. Partial parenteral nutrition was initiated. That known vaccination history, which demonstrates that severe
evening, pyrexia developed (104.6°F) as well as tachypnea disease can occur even in adult cats. Antemortem diagnos-
and increased respiratory effort. The owner elected eutha- tic testing for FPV with a fecal antigen ELISA assay or PCR
nasia. The cat regurgitated approximately 200 mL of dark assay was discussed with the owner, but was declined be-
brown fluid at the time of euthanasia. cause of the high degree of suspicion for the disease and
Gross Necropsy Findings: Ten mL of semiopaque red fluid the lack of impact that a positive or negative result would
was present in the peritoneal cavity. Streaks of hemorrhage have had on the treatment plan. Secondary bacterial sepsis
were noted throughout the skeletal muscle. Between the was suspected. Additional co-infections with other viruses,
pylorus and the ileocecocolic junction, the serosa of the such as feline coronavirus, or enteropathogenic bacteria
small intestinal tract was dark red to purple to black (see could not be ruled out. During the cat’s treatment, one of
Figure 19-2). The entire small and large intestinal tract was the kittens in the litter died suddenly and the other devel-
dilated and flaccid. The small intestinal mucosa was dark red oped signs of illness.

SUGGESTED READINGS 6. Decaro N, Desario C, Miccolupo A, et al. Genetic analysis of feline


panleukopenia viruses from cats with gastroenteritis. J Gen Virol.
Kruse BD, Unterer S, Horlacher K, et al. Prognostic factors in cats with 2008;89:2290-2298.
feline panleukopenia. J Vet Intern Med. 2010;24:1271-1276. 7. Battilani M, Balboni A, Ustulin M, et al. Genetic complexity and
Neuerer FF, Horlacher K, Truyen U, et al. Comparison of different in- multiple infections with more Parvovirus species in naturally
house test systems to detect parvovirus in faeces of cats. J Feline Med infected cats. Vet Res. 2011;42:43.
Surg. 2008;10:247-251. 8. Nakamura K, Sakamoto M, Ikeda Y, et al. Pathogenic potential of
Truyen U, Addie D, Belak S, et al. Feline panleukopenia. ABCD guidelines canine parvovirus types 2a and 2c in domestic cats. Clin Diagn Lab
on prevention and management. J Feline Med Surg. 2009;11:538-546. Immunol. 2001;8:663-668.
9. Ohshima T, Mochizuki M. Evidence for recombination between
REFERENCES feline panleukopenia virus and canine parvovirus type 2. J Vet Med
1. Verge J, Christoforoni N. La gastroenterite infectieuse des chats; est- Sci. 2009;71:403-408.
elle due a un virus filtrable. C R Seances Soc Biol Fil. 1928;99:312. 10. Ikeda Y, Mochizuki M, Naito R, et  al. Predominance of canine
2. Steinel A, Parrish CR, Bloom ME, et  al. Parvovirus infections in parvovirus (CPV) in unvaccinated cat populations and emergence
wild carnivores. J Wildl Dis. 2001;37:594-607. of new antigenic types of CPVs in cats. Virology. 2000;278:13-19.
3. Kruse BD, Unterer S, Horlacher K, et al. Prognostic factors in cats 11. Muir P, Harbour DA, Gruffydd-Jones TJ, et al. A clinical and micro-
with feline panleukopenia. J Vet Intern Med. 2010;24:1271-1276. biological study of cats with protruding nictitating membranes and
4. Addie DD, Toth S, Thompson H, et al. Detection of feline parvovi- diarrhoea: isolation of a novel virus. Vet Rec. 1990;127:324-330.
rus in dying pedigree kittens. Vet Rec. 1998;142:353-356. 12. Marshall JA, Kennett ML, Rodger SM, et al. Virus and virus-like
5. Fischer SM, Quest CM, Dubovi EJ, et al. Response of feral cats to vacci- particles in the faeces of cats with and without diarrhoea. Aust Vet
nation at the time of neutering. J Am Vet Med Assoc. 2007;230:52-58. J. 1987;64:100-105.
194 SECTION 1  Viral Diseases

13. Goodman LB, Lyi SM, Johnson NC, et al. Binding site on the trans- 28. Paltrinieri S, Crippa A, Comerio T, et al. Evaluation of inflammation
ferrin receptor for the parvovirus capsid and effects of altered affinity and immunity in cats with spontaneous parvovirus infection: conse-
on cell uptake and infection. J Virol. 2010;84:4969-4978. quences of recombinant feline interferon-omega administration. Vet
14. Moschidou P, Martella V, Lorusso E, et  al. Mixed infection by Immunol Immunopathol. 2007;118:68-74.
feline astrovirus and feline panleukopenia virus in a domestic 29. Penderis J. The wobbly cat. Diagnostic and therapeutic approach to
cat with gastroenteritis and panleukopenia. J Vet Diagn Invest. generalised ataxia. J Feline Med Surg. 2009;11:349-359.
2011;23:581-584. 30. Lappin MR, Veir J, Hawley J. Feline panleukopenia virus, feline
15. Lutz H, Castelli I, Ehrensperger F, et al. Panleukopenia-like syndrome herpesvirus-1, and feline calicivirus antibody responses in serone-
of FeLV caused by co-infection with FeLV and feline panleukopenia gative specific pathogen-free cats after a single administration of
virus. Vet Immunol Immunopathol. 1995;46:21-33. two different modified live FVRCP vaccines. J Feline Med Surg.
16. Ikegami T, Shirota K, Goto K, et al. Enterocolitis associated with 2009;11:159-162.
dual infection by Clostridium piliforme and feline panleukopenia 31. Jas D, Aeberle C, Lacombe V, et al. Onset of immunity in kittens
virus in three kittens. Vet Pathol. 1999;36:613-615. after vaccination with a non-adjuvanted vaccine against feline
17. Mochizuki M, Osawa N, Ishida T. Feline coronavirus participation panleucopenia, feline calicivirus and feline herpesvirus. Vet J.
in diarrhea of cats. J Vet Med Sci. 1999;61:1071-1073. 2009;182:86-93.
18. Sharp NJ, Davis BJ, Guy JS, et al. Hydranencephaly and cerebel- 32. Gore TC, Lakshmanan N, Williams JR, et al. Three-year duration
lar hypoplasia in two kittens attributed to intrauterine parvovirus of immunity in cats following vaccination against feline rhinotra-
infection. J Comp Pathol. 1999;121:39-53. cheitis virus, feline calicivirus, and feline panleukopenia virus. Vet
19. Url A, Truyen U, Rebel-Bauder B, et al. Evidence of parvovirus replica- Ther. 2006;7:213-222.
tion in cerebral neurons of cats. J Clin Microbiol. 2003;41:3801-3805. 33. Scott FW, Geissinger CM. Long-term immunity in cats vac-
20. Meurs KM, Fox PR, Magnon AL, et  al. Molecular screening by cinated with an inactivated trivalent vaccine. Am J Vet Res.
polymerase chain reaction detects panleukopenia virus DNA in 1999;60:652-658.
formalin-fixed hearts from cats with idiopathic cardiomyopathy 34. Day MJ, Horzinek MC, Schultz RD. WSAVA guidelines for the
and myocarditis. Cardiovasc Pathol. 2000;9:119-126. vaccination of dogs and cats. J Small Anim Pract. 2010;51:1-32.
21. Digangi BA, Gray LK, Levy JK, et al. Detection of protective antibody 35. Gamoh K, Senda M, Inoue Y, et al. Efficacy of an inactivated feline
titers against feline panleukopenia virus, feline herpesvirus-1, and panleucopenia virus vaccine against a canine parvovirus isolated
feline calicivirus in shelter cats using a point-of-care ELISA. J Feline from a domestic cat. Vet Rec. 2005;157:285-287.
Med Surg. 2011;13:912-918. 36. Chalmers WS, Truyen U, Greenwood NM, et al. Efficacy of feline
22. Abd-Eldaim M, Beall M, Kennedy M. Detection of feline panleu- panleucopenia vaccine to prevent infection with an isolate of
kopenia virus using a commercial ELISA for canine parvovirus. Vet CPV2b obtained from a cat. Vet Microbiol. 1999;69:41-45.
Ther. 2009;10:E1-E6. 37. Nakamura K, Ikeda Y, Miyazawa T, et  al. Characterisation
23. Neuerer FF, Horlacher K, Truyen U, et al. Comparison of different of cross-reactivity of virus neutralising antibodies induced by
in-house test systems to detect parvovirus in faeces of cats. J Feline feline panleukopenia virus and canine parvoviruses. Res Vet Sci.
Med Surg. 2008;10:247-251. 2001;71:219-222.
24. Patterson EV, Reese MJ, Tucker SJ, et  al. Effect of vaccination 38. Decaro N, Buonavoglia D, Desario C, et  al. Characterisation of
on parvovirus antigen testing in kittens. J Am Vet Med Assoc. canine parvovirus strains isolated from cats with feline panleuko-
2007;230:359-363. penia. Res Vet Sci. 2010;89:275-278.
25. Decaro N, Desario C, Lucente MS, et al. Specific identification of 39. Truyen U, Addie D, Belak S, et  al. Feline panleukopenia. ABCD
feline panleukopenia virus and its rapid differentiation from canine guidelines on prevention and management. J Feline Med Surg.
parvoviruses using minor groove binder probes. J Virol Methods. 2009;11:538-546.
2008;147:67-71. 40. Levy JK, Crawford PC, Collante WR, et al. Use of adult cat serum
26. Horiuchi M, Yuri K, Soma T, et al. Differentiation of vaccine virus to correct failure of passive transfer in kittens. J Am Vet Med Assoc.
from field isolates of feline panleukopenia virus by polymerase 2001;219:1401-1405.
chain reaction and restriction fragment length polymorphism anal- 41. Yang S, Wang S, Feng H, et  al. Isolation and characterization of
ysis. Vet Microbiol. 1996;53:283-293. feline panleukopenia virus from a diarrheic monkey. Vet Microbiol.
27. De Lahunta A. Comments on cerebellar ataxia and its congenital 2010;143:155-159.
transmission in cats by feline panleukopenia virus. J Am Vet Med
Assoc. 1971;158(Suppl 2):901-906.
CHAPTER 20

Feline Coronavirus Infection


Jane E. Sykes

3 months to 3 years of age, with at least 50% of affected cats


Overview of Feline Coronavirus Infections aged 12 months or younger (Figure 20-1). However, FIP can
First Described: 19631; a viral etiology was not identified until occur at any age, and there is a secondary peak of incidence
the 1970s. in geriatric cats (>10 years of age), possibly as a result of sub-
Cause: Feline coronavirus (Family Coronaviridae, genus optimal immune function. Males and sexually intact cats have
Coronavirus) been predisposed in some studies,9-11 and a disease peak may
exist in the fall and winter.12 Although the disease occurs in
Affected Hosts: Cats and wild felids, especially cheetahs
all breeds, purebred cats are more susceptible; Abyssinians,
Mode of Transmission: Fecal-oral Australian mist, Bengals, birmans, Burmese, British shorthairs,
Geographic Distribution: Worldwide Himalayans, ragdolls, rexes, and possibly Scottish folds may
be predisposed.9-14 Breed predispositions may vary geographi-
Major Clinical Signs: Fever, lethargy, inappetence, vomiting, cally and temporally depending on the preferences of breeders
diarrhea, dehydration, icterus, tachypnea, uveitis, neuro- in a region, and specific lines may be more predisposed than the
logic signs, abdominal distention due to ascites. breeds themselves.6 The molecular basis of genetic susceptibility
Differential Diagnoses: Toxoplasmosis, congestive heart to FIP is currently unclear. Siblings of cats that die of FIP may
failure, carcinomatosis, lymphoma, pancreatitis, rabies, be at increased risk for FIP.15
cryptococcosis, bacterial peritonitis, pyothorax, bacterial In multiple-cat household situations, cats are repeatedly
meningitis, chronic stomatitis, multiple myeloma, infec- infected, shed virus, and recover, but some cats remain persis-
tion with FeLV or FIV. tently infected and chronically shed FCoV in the absence of clini-
Human Health Significance: Feline coronaviruses do not cal signs (Figure 20-2). More than half, and as many as 100% of
infect humans. cats in environments with more than six cats, become infected
with FCoVs.6 The seroprevalence is lower in cats from single-cat
households and among feral cats.16 However, even though the
prevalence of infection in multicat households is high, fewer than
Etiology and Epidemiology 10% of cats from large, multicat households ultimately develop
FIP. Thus, although the incidence of infection is high, the inci-
Coronaviruses are large, enveloped, single-stranded RNA dence of disease in single- or two-cat households is only around
viruses with club-shaped spikes on their outer surface (see 1 in 5000; in catteries it is around 5% to 10%.17,18 Provided
­Figure 14-1, B). They have the largest RNA genomes of all they are unrelated by birth, cats in households with a history of
known viruses. Feline coronaviruses (FCoV), like canine enteric FIP are not more likely to develop FIP than cats in households
coronavirus, belong to the Group 1a coronaviruses (see Box without FIP.19 Thus, FIP is usually a sporadic disease that does
17-1). In fact, even canine enteric coronavirus has the poten- not spread from one cat to another. However, every few years,
tial to infect cats and cause diseases similar to those caused by epidemics of disease can occur in catteries or shelters, with mor-
FCoV.2 Among FCoVs, there are two different serotypes, type tality rates that exceed 10%.12 Because it is an enveloped virus,
I and type II, which use different receptors for cellular entry FCoV is readily inactivated by disinfectants and generally sur-
in  vitro3,4 but cause the same clinical manifestations. Type I vives less than a day or two at room temperature. However, the
strains predominate worldwide.5-7 Type II strains, which are possibility of prolonged survival (up to 7 weeks) in the environ-
thought to have evolved from genetic recombination between ment under certain conditions has been suggested.15,20 In this
canine enteric coronavirus and FCoV, are more readily grown situation, fomites might play an important role in transmission.
in culture and so have been more extensively studied; they The epidemiology and pathogenesis of FIP has both fascinated
possess a spike protein that resembles that of canine enteric and confused veterinary virologists worldwide for decades. The
coronavirus. most widely accepted theory (the “internal mutation hypothe-
FCoVs cause enteric disease in cats as well as feline infec- sis”) is that cats are initially infected with a low-pathogenicity
tious peritonitis (FIP), a serious systemic pyogranulomatous to coronavirus after oronasal exposure, which results either in no
granulomatous disease that progresses over a period of weeks to signs, or mild enteric disease. This low-pathogenicity virus has
months and, once it occurs, is ultimately always fatal. FIP is a been referred to as feline enteric coronavirus in some publica-
major cause of death in young and young adult cats, especially tions in order to distinguish it from virulent FIP virus. The use
cats from multicat environments such as purebred catteries of this name has been controversial, because although the virus
and shelters. Wild cats, especially cheetahs, are also suscepti- is primarily confined to the gastrointestinal tract (and especially
ble.8 The vast majority of domestic cats that develop FIP are colonic epithelial cells), FCoV RNA can also be found in blood

195
196 SECTION 1  Viral Diseases

30 FCoV strains. However, mutations in the spike protein gene,24,25


membrane protein gene,26 and the nonstructural 3c and 7b
genes23,27-29 may play a role. In particular, the 3c gene appears to
be disrupted in many (but not all) virulent FCoV strains.
Number of cats

20
The other hypothesis proposed to explain the pathogenesis
of FIP is that distinct circulating virulent and avirulent FCoV
strains exist, and the combination of infection with a virulent
10
FCoV and an individual cat’s genetic and environmental pre-
dispositions leads to FIP.25 It has also been suggested that both
hypotheses may play a role.30
0
3-6 7-12 13-24 25-36 37-60 61-120 120-180 >180 Clinical Features
Age (months)
FIGURE 20-1  Age distribution of 99 cats with necropsy-confirmed FIP at the UC Signs and Their Pathogenesis
Davis VMTH. An additional six cats were reported to be “juvenile” or kittens. There were 38 Cats are usually infected with FCoV by oronasal exposure
females (17 intact) and 66 males (22 intact). to virus in feces or fomites contaminated with fecal material.
Shared litter boxes are thought to play a major role in transmis-
sion.15 Replication of low-pathogenicity strains of FCoV in epi-
thelial cells at the tips of intestinal villi may be associated with
Overcrowded environment no signs, or acute or chronic, persistent or intermittent small-
Stress, fecal contamination bowel diarrhea, and less commonly, vomiting and/or inappe-
tence. Transient upper respiratory signs have been reported in
some cats on initial infection with FCoV.15 Virus is shed in the
feces from 1 week after infection. Some cats then shed large
quantities of virus continuously for life.12,22,31-33
Both serotype I and serotype II strains appear to enter macro-
phages via a lectin receptor known as fDC-SIGN (feline dendritic
cell-specific intercellular adhesion molecule grabbing non-inte-
grin receptor).3,34 Replication of virulent FCoV strains within
macrophages results in two forms of disease, which reflect the
immune response mounted by the host. FIP is an immune com-
plex disease. Noneffusive (“dry”) FIP occurs in cats that mount
a partial CMI response and is characterized by pyogranuloma-
tous to granulomatous inflammation within a variety of organs,
Genetically-predisposed Chronic coronavirus but especially the mesenteric lymph nodes, kidneys, liver, lungs,
cats shedders brain, and eye. Solitary or multifocal granulomas of the intesti-
FIGURE 20-2  Interplay between genetics, virus shedding, and environment in feline nal wall also occasionally develop, especially in the region of the
coronavirus infections and FIP. ileocecal junction (Figure 20-3).15 Effusive (“wet”) FIP occurs in
cats that are unable to mount an immune response and is char-
acterized by accumulation of high protein exudates in the thorax
and tissue macrophages of cats that do not have FIP.21,22 In some and/or abdomen, which typically contain low numbers of cells.
infected cats, the low-pathogenicity virus is believed to mutate to Production of vascular endothelial growth factor by infected
a virulent strain that can multiply within macrophages without monocytes may be lead to increased vascular permeability and
hindrance by the immune system and incite a systemic pyogranu- contribute to cavitary effusion.35 Many cats have a mixture of
lomatous vasculitis. The mutation may occur shortly after initial both forms of the disease, and noneffusive disease may prog-
infection, or years later, which may explain why some indoor cats ress to effusive disease. Infection itself results in immune dys-
from single-cat households develop FIP several years after they regulation, with a profound, virus-induced depletion of CD4+
are acquired. Virulent strains may not be able to replicate effec- and CD8+ cells; production of TNF-α, granulocyte-macrophage
tively within the gut,23 which may be the reason why cat-to-cat colony stimulating factor (GM-CSF), and granulocyte colony-
transmission of FIP does not occur, yet the disease can be trans- stimulating factor (G-CSF) by infected macrophages; impaired
mitted effectively by inoculating naïve cats with effusion from IFN-γ production; and hypergammaglobulinemia.36-38 The
a cat with FIP. Factors that contribute to immunosuppression, mechanism of T-cell depletion is not clear, as the virus does not
such as concurrent viral infection, stress due to overcrowding, infect lymphocytes, only monocytes and macrophages. Infection
surgery, or transport, and especially genetic factors may allow of antigen-presenting cells, specifically dendritic cells, has been
viral replication and mutation to proceed unchecked. Simulta- hypothesized to lead to T-cell apoptosis. Progressive immune
neous immune compromise of a large number of cats, such as system failure may be associated with a conversion to predomi-
in a shelter situation, may explain epidemics of FIP. Other risk nantly effusive disease manifestations. Despite the profound
factors for FIP include regular introduction of new cats to a cat- T-cell deficiency that accompanies FIP, opportunistic infections
tery and the proportion of cats in a cattery that shed coronavirus are rarely reported. Nevertheless, concurrent infections with
chronically.12 There is no distinct mutation that allows avirulent retroviruses and Toxoplasma gondii and opportunistic bacterial
FCoV strains to be differentiated from virulent strains, and there- infections can occur;6 the author is aware of one cat that was
fore no diagnostic test exists that distinguishes FIP from benign co-infected with Sporothrix schenckii.
CHAPTER 20  Feline Coronavirus Infection 197

FIGURE 20-3  Colonic mass removed at surgery from a 7-month-old female spayed
domestic shorthair with anorexia and hematochezia. Histopathology showed severe, mul-
tifocal coalescing pyogranulomatous colitis and lymphadenitis. (Courtesy of the University
of California, Davis Veterinary Anatomic Pathology service.) FIGURE 20-5  Keratic precipitates in a 5-year-old intact male Burmese cat with FIP.
(Courtesy of the University of California, Davis Veterinary Ophthalmology service.)

neoplasia.39 Rarely, pericardial effusion results in cardiac tam-


ponade. Pyogranulomatous or granulomatous inflammation
may lead to mesenteric lymphadenomegaly, irregular renomeg-
aly, intestinal masses, hepatomegaly, icterus, pneumonia, uve-
itis, chorioretinitis, and, rarely, nodular skin lesions. Neurologic
signs, which can include focal or generalized seizures, occur in
at least 10% of cats with FIP and result primarily from menin-
goencephalitis, meningomyelitis, ependymitis, choroiditis, and
obstructive hydrocephalus. Obstructive hydrocephalus occurs
FIGURE 20-4  Obstructive hydrocephalus in an 8-month-old male neutered exotic secondary to choroiditis and ependymitis (Figure 20-4). In one
shorthair cat that developed ataxia and head tremors. Hydrocephalus and secondary study, FIP was responsible for almost half of all neurologic
cerebellar herniation were found at necropsy. Histopathology revealed severe, multifocal
disease in 97 cats due to infectious or inflammatory causes.40
pyogranulomatous meningoencephalitis, choroiditis, and ventriculitis, and pyogranulo-
matous inflammatory lesions were also found throughout the thoracic and abdominal Occasionally profound anemia occurs secondary to immune-
viscera. (Courtesy of the University of California, Davis Veterinary Anatomic Pathology mediated hemolysis13,14 or possibly microangiopathic damage,
service.) whereby erythrocytes are lysed as they travel through inflamed
blood vessels. Immune-mediated glomerulonephritis has also
been reported, and FIP should always be considered in cats with
The incubation period for FIP is highly variable. Kittens usu- protein-losing nephropathy, which is otherwise rare in cats.41
ally become infected at 4 to 8 weeks of age, when maternal Uncommonly, lameness occurs as a result of synovitis.6
antibody begins to wane, but infections have been reported in
kittens as young as 2 weeks of age.20 Disease may occur a few Physical Examination Findings
weeks after infection or years later, but most often it occurs 6 Physical examination findings in cats with FIP reflect the type
to 18 months after initial infection.19 Even after the onset of of disease present (effusive versus noneffusive) and the loca-
systemic pyogranulomatous inflammatory disease, clinical signs tion where lesions occur. Cats with respiratory tract involve-
may not be apparent for months. In support of this, lesions ment may show tachypnea, and if there is pleural effusion, a
consistent with FIP have been found incidentally in cats during rapid, shallow breathing pattern and muffled heart and lung
abdominal surgery such as ovariohysterectomy.20 sounds may be present. Other signs include pyrexia, dehydra-
The clinical signs of FIP often change over time and depend tion, mucosal pallor or icterus, a thin body condition, and
on the organs affected and the relative predominance of inflam- evidence of ascites. Abdominal palpation may reveal hepato-
matory versus effusive disease manifestations. The most com- megaly, irregular renomegaly, and/or abdominal mass lesions
mon signs are lethargy and inappetence, as well as a fluctuating that result from mesenteric lymphadenomegaly or intestinal
fever that does not respond to antibacterial drug treatment. pyogranulomas. Sometimes pain is appreciated on abdominal
Nevertheless, many cats are bright, appetent, and in good body palpation, which may reflect pancreatic involvement in some
condition early in the course of illness. Some cats have increased cats. Testicular enlargement may be detected in intact male
thirst and urination, possibly secondary to pyrexia. Ultimately, cats. A wide range of neurologic signs may be present, such as
weight loss develops, but owners of cats that develop abdomi- obtundation, twitching, tremors, behavioral changes, nystag-
nal distention may mistake the distention for weight gain or mus, hyperesthesia, exaggerated segmental reflexes, ataxia, uri-
pregnancy. Stunted growth may occur in affected kittens. Pleu- nary incontinence, or cranial nerve defects. Ocular signs include
ral effusion may be associated with tachypnea and respiratory conjunctivitis, mucopurulent ocular discharge, thickening and
distress. Testicular enlargement may occur in cats with serositis hyperemia of the nictitans, uveitis with dyscoria or anisoco-
that involves the tunica vaginalis. FIP is responsible for approxi- ria, aqueous flare, keratic precipitates, hypopyon, hyphema,
mately 10% of pericardial effusions in cats, the third most com- chorioretinitis, perivascular infiltrates, retinal detachment, or
mon cause of pericardial effusion after cardiomyopathy and blindness (Figure 20-5).
198 SECTION 1  Viral Diseases

TABLE 20-1
Complete Blood Count Findings at Admission in 38 Cats with Necropsy-Confirmed Feline Infectious Peritonitis
at the UC Davis VMTH
Percent below Percent within Percent above Range for
Reference the Reference the Reference the Reference Cats with Number
Test Range Range Range Range FIP Tested
Hematocrit (%) 30-50 68 32 0 17-53 38
MCV (fL) 65-75 18 82 0 36-52 38
MCHC (g/dL) 33-36 5 58 37 28-36 38
RDW (%) 14-18 0 30 70 14-33 27
Neutrophils* 2000-9000 3 26 71 416-49,313 38
(cells/µL)
Band neutrophils* 0-rare 0 50 50 0-3251 38
(cells/µL)
Metamyelocytes 0 0 95 5 0-276 38
(cells/µL)
Monocytes 50-600 5 71 24 0-820 38
(cells/µL)
Lymphocytes 1000-7000 58 42 0 89-6886 38
(cells/µL)
Eosinophils 150-1100 71 29 0 0-770 38
(cells/µL)
Platelets 180,000-500,000 37 44 19 30,000-874,000 27†
(cells/µL)

FIP, Feline infectious peritonitis; RDW, red cell distribution width.


*22 (58%) had evidence of toxic neutrophils.
†A smear was evaluated manually for 37 of the 38 cats. The presence of macroplatelets were reported for 18 (49%) of cats.

Diagnosis regenerative or nonregenerative (Table 20-1). Microcytosis may


be present. Examination of erythrocyte morphology occasion-
Currently, definitive diagnosis of FIP is made only by immunohis- ally reveals schistocytosis, mild normoblastosis, or agglutina-
tochemical staining for coronavirus antigen within lesions charac- tion. There may be a leukocytosis due to a neutrophilia and
terized by pyogranulomatous or granulomatous vasculitis. Because monocytosis, or leukopenia. Lymphopenia occurs in more than
it can be difficult or impossible to safely obtain biopsy specimens 50% of affected cats, and eosinopenia is also common. In some
from cats with FIP, antemortem diagnosis is often only suspected cats, a left shift and evidence of toxic neutrophils are seen. Mild
on the basis of history, signalment, and clinical and laboratory to moderate thrombocytopenia is common in cats with noneffu-
findings, and by ruling out other causes of disease. Provided it is sive disease and may reflect the presence of disseminated intra-
correctly performed and interpreted, immunocytochemistry may vascular coagulation or immune-mediated platelet destruction.
be helpful. Because the presence of the characteristic effusion is However, thrombocytosis can also occur.
most helpful for antemortem diagnosis, efforts should be always
made to identify and analyze any fluid that is present in body cavi- Serum Biochemical Tests
ties. When owner funds are limited, laboratory analysis of effu- Many cats with FIP have hyperproteinemia due to hyper-
sion, rather than blood, may be the most economic diagnostic globulinemia, which results from a polyclonal gammopa-
approach. Unfortunately, the lack of a definitive noninvasive diag- thy (Figure 20-6). Rarely, a monoclonal gammopathy can
nostic assay for FIP and the extremely poor prognosis sometimes occur.42 Total protein concentrations may be as high as 12 g/
leads clinicians to perform large numbers of diagnostic tests in the dL (Table 20-2).20 In one study, hyperglobulinemia was pres-
hope that an answer will appear. In other situations, the diagnosis ent in 50% of cats with effusion and 70% of cats without
of FIP is made too hastily, and euthanasia is performed without effusion.43 Globulin concentration may decrease terminally,
sufficient clinical and laboratory justification. so cats with advanced disease may have protein concentra-
tions that are within the reference range.14 Hypoalbumin-
Laboratory Abnormalities emia is often present because of liver involvement, leakage
Complete Blood Count from damaged vessels, urinary loss in cats with glomerulone-
A mild, nonregenerative anemia is often present in cats with FIP, phritis, or inflammation (albumin is a negative acute-phase
and sometimes severe anemia occurs, which is usually poorly reactant protein). Thus, the serum albumin:globulin ratio
CHAPTER 20  Feline Coronavirus Infection 199

A B
FIGURE 20-6  A, Densitometric scan of serum protein electrophoresis of normal feline serum. B, Scan from a 9-month-old male neutered domestic shorthair cat with FIP. There is a
polyclonal gammopathy, represented by a broad peak in the γ-globulin region, with a mild decrease in the albumin and mild increases in the α2 and β1 fractions. (A redrawn from Baker
RJ, Valli VE. Electrophoretic and immunoelectrophoretic analysis of feline serum proteins. Am J Vet Res 1988;52[3]:308-304.)

TABLE 20-2
Findings on Serum Biochemistry Analysis in 36 Cats with Necropsy-Confirmed Feline Infectious Peritonitis
at the UC Davis VMTH
Percent below Percent within Percent above
Reference the Reference the Reference the Reference Range for Number of
Test Range Range Range Range cats with FIP Cats Tested
Sodium (mmol/L) 151-158 94 6 0 129-152 35
Potassium (mmol/L) 3.6-4.9 26 74 0 2.2-5.4 35
Chloride (mmol/L) 117-126 91 9 0 94-121 35
Bicarbonate (mmol/L) 15-21 8 75 17 12-25 36
Calcium (mg/dL) 9.0-10.9 56 44 0 6.6-10.6 36
Phosphorus (mg/dL) 3.2-6.3 8 69 22 1.9-8.4 36
Creatinine (mg/dL) 1.1-2.2 69 28 3 0.4-2.8 36
BUN (mg/dL) 18-33 56 31 14 10-58 36
Glucose (mg/dL) 63-118 0 47 53 63-381 36
Total protein (g/dL) 6.6-8.4 31 28 42 4.1-11.9 36
Albumin (g/dL) 2.2-4.6 50 50 0 0.9-3.6 36
Globulin (g/dL) 2.8-5.4 3 42 56 2.5-9.4 36
Cholesterol (mg/dL) 89-258 17 83 0 56-247 36
Total bilirubin (mg/dL) 0-0.2 0 40 60 0-5.3 36
ALT (U/L) 27-101 50 25 25 18-648 36
AST (U/L) 17-58 6 43 51 0-1554 36
ALP (U/L) 14-71 28 61 11 0-161 36
GGT (U/L) 0-4 0 96 4 0-5 23

FIP, feline infectious peritonitis.

may be more useful than the globulin alone for diagnosis; not clear, but it may result from hemolysis, hepatic necrosis,
ratios less than 0.8 are uncommon (but not impossible) in and/or cholestasis.
cats with FIP, so they help to rule out (but not to rule in) a Measurement of α1-acid glycoprotein (an acute phase pro-
diagnosis of FIP.44,45 Other variable findings include hypona- tein) has been suggested for diagnosis, because serum con-
tremia, hypokalemia, hypochloremia, hyperglycemia, azote- centrations often exceed 1500 µg/mL in cats with FIP.20,46,47
mia, increased liver enzyme activities, hypocholesterolemia, However, α1-acid glycoprotein concentrations also increase
and hyperbilirubinemia. The cause of hyperbilirubinemia is with other inflammatory diseases.20
200 SECTION 1  Viral Diseases

TABLE 20-3 TABLE 20-4


Composition of Body Cavity Effusions from 21 Cats with Composition of Cerebrospinal Fluid from 10 Cats with Necropsy-
Necropsy-Confirmed Feline Infectious Peritonitis at the Confirmed Feline Infectious Peritonitis at the UC Davis VMTH
UC Davis VMTH
Number
Number Reference of Cats
of Cats Test Range Median Range Tested
Test Range Mean ± SD Tested Total protein 44-4079 639 <25 4*
Total protein (g/dL) 2.9-8.1 5.1 ± 1.8 21 (mg/dL)
RBC (cells/µL) <100-38,600 ND 18 RBC (cells/µL) 3-850 340 0 9
TNC (cells/µL) 200-13,200 3683 ± 3474 19 TNC (cells/µL) 26-2637 303 0-2 10
Neutrophils (%) 3-97 61 ± 28 21 Neutrophils (%) 4-90 73 10
Lymphocytes (%) 0-22 6±6 21 Lymphocytes (%) 5-89 19 10
Monocytes (%) 1-96 33 ± 26 21 Monocytes (%) 0-20 7 10

Nineteen specimens were abdominal and two were pleural effusions. TNC, total neutrophil count.
ND, Not determined; SD, standard deviation; TNC, total neutrophil *Insufficient quantity available from some cats for determination of
count. protein concentration.

Urinalysis younger cats, the positive predictive value of the test is higher,
The urinalysis in cats with FIP may be unremarkable or contain because diseases such as lymphoma and bacterial peritonitis are
protein due to glomerular or tubular damage. Hematuria and, less common. Positive test results indicate only the presence of
less commonly, pyuria and cylindruria may be present. Bilirubi- an exudate, so cytologic examination of the fluid must still be
nuria may be detected in cats with liver injury. performed.

Coagulation Profile Cerebrospinal Fluid Analysis


In addition to thrombocytopenia, abnormalities of coagula- The cerebrospinal fluid (CSF) of cats with neurologic FIP often
tion in cats with FIP include prolonged prothrombin time has increased protein content (30 to more than 1000 mg/dL, ref-
and partial thromboplastin time as a result of severe liver erence range less than 25 mg/dL) and increased total nucleated
injury, and increased fibrin degradation product or D-dimer cell count (20 to 10,000 cells/µL), usually consisting of a mixed
concentrations.15 but predominantly neutrophilic cellular pleocytosis (Table
20-4). In some cats, protein content and leukocyte counts are
Analysis of Effusion Fluid normal.51,52
The “classic” FIP effusion fluid is a high-protein (greater than
3.5 g/dL) exudate that contains a low number of nucleated Diagnostic Imaging
cells (<5000 cells/µL), usually nondegenerate to mildly degen- Plain Radiography
erate neutrophils and macrophages (Table 20-3). Erythropha- Plain thoracic radiography may reveal pleural effusion, enlarge-
gocytosis, leukophagia, and reactive mesothelial cells can be ment of the cardiac silhouette in cats with pericardial effusion,
observed in the fluid from some cats. Grossly, the fluid has a and pulmonary nodular or peribronchial infiltrates in cats with
yellow appearance and may contain fibrin clots. However, the pyogranulomatous pneumonia (Figure 20-7). Abdominal radio-
total protein content and cell counts of abdominal and pleural graphs may show loss of peritoneal or retroperitoneal detail due
effusions vary considerably, which complicates the diagnosis for to peritoneal effusion, hepatomegaly, splenomegaly, renomeg-
some cats with effusive disease. Very rarely, chylous effusions aly, or mass lesions associated with the gastrointestinal tract or
occur.48 An effusion albumin/globulin ratio below 0.4 is sug- abdominal lymph nodes.
gestive of FIP.49
The Rivalta test is a simple test that can differentiate between Sonographic Findings
transudates and exudates. In this test, a drop of 98% glacial Abdominal ultrasound findings in FIP include the presence of
acetic acid is mixed with 7 to 8 mL of distilled water in a trans- anechoic or mildly echogenic peritoneal fluid; hyperechogenicity
parent 10-mL tube. A drop of effusion is then added to the tube, and “clumping” of the mesentery; enlarged and hypoechoic
and if it dissipates in the solution, the test is negative. If it retains abdominal lymph nodes (Figure 20-8, A); enlargement and dif-
its shape, stays attached to the surface, or moves slowly down fuse or focal hypoechogenicity of the liver and spleen;53 renal
in the solution, then the test is positive.20 In a study of cats asymmetry with increased cortical echogenicity, hypoechoic
with effusion, 35% of which had FIP and a conclusive Rivalta nodules, subcapsular fluid accumulation, or loss of corticome-
test, the positive predictive value of this test for the diagnosis dullary distinction (see Figure 20-8, B); and/or thickening of all
of FIP was 58% (58% chance that a cat that tests positive truly intestinal wall layers or intestinal mass lesions. Pleural effusion
has FIP), and the negative predictive value was 93% (93% or comet-tail artifacts (due to pulmonary infiltrates) may be seen
chance that a cat that tests negative does not have FIP).50 In through the diaphragm.
CHAPTER 20  Feline Coronavirus Infection 201

FIGURE 20-7  Lateral thoracic radiograph from a 9-month-old male neutered A


domestic shorthair cat with FIP and pyogranulomatous pneumonia. There is a severe, dif-
fuse, patchy alveolar and nodular interstitial pattern with thickening of the bronchial walls
and mild pleural effusion.

Magnetic Resonance Imaging of the Central Nervous System


Findings on MRI that suggest FIP consist of ventricular dilata-
tion and variable contrast enhancement of the periventricular
regions, choroid, and meninges. In some cats, MRI findings are
unremarkable.

Microbiologic Tests
Serologic Diagnosis
Detection of antibodies to FCoV can be performed using immu-
nofluorescent antibody testing, ELISA, or virus neutralization.54 B
The methods used, as well as the titers themselves, vary con- FIGURE 20-8  A, Abdominal ultrasound image from a 9-month-old male neutered
siderably between laboratories. For example, some laboratories domestic shorthair cat with FIP and ileocecocolic lymphadenopathy. The lymph nodes are
use related coronaviruses as a source of antigen for the test, enlarged and hypoechoic. B, Abdominal ultrasound image from a 1-year-old male intact
rather than FCoV.15 Use of a reliable laboratory that reports Scottish fold with FIP. There is renal irregularity and subcapsular fluid, as well as moderate
quantitative titers (to the endpoint dilution, as well as down peritoneal effusion.
to 1:100) is critical. Even when performed correctly, a posi-
tive FCoV antibody titer is not diagnostic for FIP, because cats
that have been exposed to avirulent FCoV strains or even other antibody in the CSF correlated well with a diagnosis of FIP in
related coronaviruses are also seropositive. Therefore, serology one study,52 but not in another study.51 In addition, the pres-
is a “coronavirus antibody test” and not an “FIP test.” It has ence of sufficient quantities of CSF for serology are frequently
been suggested that more cats have been killed as a result of not available.
misinterpretation of FCoV antibody tests than by the disease
itself.20 Certainly a diagnosis of FIP should never be made based Molecular Diagnosis Using the Polymerase Chain Reaction
on the presence of nonspecific clinical or laboratory abnor- Real-time reverse transcriptase–PCR (RT-PCR) assays have
malities such as fever or leukocytosis and a positive coronavi- been developed for detection of FCoV, but these do not differ-
rus antibody test. Occasionally (up to 10% of the time), cats entiate between virulent and avirulent strains. In addition, avir-
with advanced disease are seronegative, because of failure of ulent strains can be found in the blood and tissues of cats that
antibody production with severe immunosuppression, or the do not have FIP,21,22 so the finding of virus in locations other
complexing of antibody by the large quantities of virus pres- than the gastrointestinal tract is not helpful for diagnosis. False-
ent. In one study, titers of 1:1600 or higher were highly sugges- negative test results can occur when there are low quantities
tive (94% chance) of FIP in the presence of compatible clinical of virus present or if degradation of RNA occurs during speci-
signs.44 In addition, strong positive titers (e.g., ≥ 1:6400) in cats men transport. Some RT-PCR assays do not detect all strains of
with consistent signs and laboratory abnormalities support a FCoV. Positive RT-PCR results in blood or effusion fluid from
diagnosis of FIP if a cat resides in a household that contains cats with other clinical abnormalities that suggest FIP do indi-
only one or two cats, because cats often become seronegative cate the presence of a coronavirus and, in that respect, may help
within a few months once they are removed from households to support the diagnosis made, provided the limitations of the
that contain large numbers of cats. test are recognized.
Other body fluids can also be analyzed for antibodies to
FCoV. In one study, positive antibody titers in effusion had Immunostaining of FCoV Antigen
a positive predictive value of 90% and a negative predictive FCoV antigen can be detected in macrophages with immu-
value of 79%, but the magnitude of the titer did not corre- nocytochemistry or immunohistochemistry (Figure 20-9).
late with the diagnosis of FIP.44 The presence of anti-FCoV Either fluorescent antibody or immunoperoxidase methods
202 SECTION 1  Viral Diseases

B
FIGURE 20-9  A, Cytospin preparation of a tracheobronchial lavage from the cat in Figure 20-7. The specimen was highly cellular (4400 cells/µL) and the cells consisted of 42% degen-
erate and nondegenerate neutrophils, 5% lymphocytes, and 53% macrophages. B, Immunocytochemistry stain on the same specimen for coronavirus antigen showing positive staining
in association with macrophages.

may be used (Table 20-5). When antigen tests are positive, may be clumped. Abdominal organs may be enlarged or irreg-
provided the test is performed and interpreted properly (with ular. Granulomas appear as variably sized multifocal white,
use of positive and negative control slides), studies suggest cream, tan, or yellow nodular lesions on serosal surfaces and
that only cats with FIP have positive test results.44,55 False- within the parenchyma of organs such as the lungs, spleen, kid-
negative results occur when there are insufficient numbers of neys, pancreas, and liver (see Figure 20-10, B). Lesions have also
infected cells, when low quantities of virus are present, or been described within the nasal cavity and sinuses. Pyogranulo-
when antigen is unavailable for detection because of complex- mas may be visible grossly as miliary lesions, or they may be
ing by antibody. several centimeters in diameter. Thoracic and/or abdominal
lymphadenomegaly is a common finding. Diffuse or focal thick-
Pathologic Findings ening of the intestinal wall or intestinal mass lesions may be
Gross Pathologic Findings present. Examination of the brain can reveal fibrinous exudate
At necropsy, gross findings in cats with FIP include variable in association with the meninges, with or without ventricular
quantities of pleural, pericardial, and peritoneal effusion (Figure dilation and hydrocephalus (see Figure 20-4). Thymic involu-
20-10, A). Fibrin adhesions may be present and the mesentery tion may also be present.
CHAPTER 20  Feline Coronavirus Infection 203

TABLE 20-5
Assays Available for Diagnosis of Feline Infectious Peritonitis
Assay Specimen Type Target Performance
Fluorescent or Wash or effusion FCoV Gold standard for diagnosis. False negatives can occur in
immunoperoxidase specimens, tissue specimens that contain low numbers of macrophages or virus
antibody staining aspirates, tissues particles, or when virus is complexed by antibody. Immuno-
obtained at biopsy fluorescence is more sensitive than immunoperoxidase methods.
or necropsy Non-specific staining may be interpreted as positive results by
untrained personnel.
Serology Blood, CSF, aqueous FCoV antibody Positive antibody titers reflect only antibodies to a coronavirus
humor, effusion and are not specific for a diagnosis of FIP. Most cats in multicat
fluid households test positive. Negative titers can occur in cats with
advanced FIP. High titers in cats that do not reside in multicat
households and that have signs suggestive of FIP may support
the diagnosis. Interlaboratory variation in methodology and
titer reporting occurs.
RT-PCR Blood, wash or FCoV RNA Does not differentiate between virulent and avirulent FCoV
­effusion specimens, strains, and avirulent strains may be found in tissues and blood.
tissue aspirates, Sensitivity and specificity can vary depending on assay design.
tissues obtained at False negative results occur when virus levels are low, when
biopsy or necropsy variant virus strains are present, or as a result of degradation of
viral nucleic acid during specimen transport.
Histopathology Usually necropsy Inflammatory Biopsy is often not feasible antemortem as a result of critical ill-
specimens, but also ­lesions induced ness and coagulopathies.
biopsies by FCoV (pyo-
granulomatous
vasculitis)

FCoV, feline coronavirus; FIP, feline infectious peritonitis; RT-PCR, reverse transcriptase–polymerase chain reaction.

Histopathologic Findings recombinant IFN-α, but none have convincingly shown ben-
The characteristic histopathologic findings of FIP are systemic efit in vivo. Prolonged remissions were reported in several cats
perivascular, multifocal to coalescing pyogranulomatous or treated with a combination of glucocorticoids and feline IFN-
granulomatous inflammatory lesions (Figure 20-11). Lesions ω,56 but a randomized, placebo-controlled clinical trial reported
predominantly contain macrophages and neutrophils, with no effect of feline IFN-ω.57 Other drugs used to treat cats with
lesser numbers of lymphocytes and plasma cells, although occa- FIP include the immunomodulatory drugs pentoxifylline and
sionally the histiocytic or the lymphoplasmacytic component Polyprenyl Immunostimulant (see Chapter 7)58; ozagrel hydro-
of the inflammatory response is more florid. Necrosis may be chloride, a thromboxane synthetase inhibitor59; and the antivi-
present within the lesions. Lesions in the central nervous sys- ral drug nelfinavir.14,60 Controlled clinical trials are required to
tem consist of pyogranulomatous meningoencephalomyelitis assess the efficacy and safety of these treatments. Because FIP is
and choroiditis. Other findings that may be identified include an immune-mediated disease, nonspecific immune stimulation
lymphoid depletion, which results from apoptosis, and mem- has the potential to cause harm. The use of small interfering
branous glomerulonephritis. RNA molecules, which bind viral RNA and prevent viral repli-
cation, has recently shown promising results in vitro.61 Cyclo-
Treatment and Prognosis sporin inhibits FIPV replication in vitro;62 studies are required
to determine if cyclosporin treatment benefits infected cats or
Currently, no cure for FIP exists; it is a progressive, invari- whether harm results from immunosuppression.
ably fatal disease. The goal of treatment is to prolong life span Supportive treatments that may be required include subcu-
and improve quality of life through reduction of inflamma- taneous fluid therapy and nutritional support. Inappetent cats
tion and supportive care. The most effective treatment known can benefit from enteral nutrition through a feeding tube. The
is prednisolone, administration of which results in temporary use of broad-spectrum antimicrobial drugs to treat cats with FIP
remissions in some cats (Table 20-6). Other immunosuppres- is controversial; it may only promote opportunistic infections
sive drugs, such as chlorambucil and cyclophosphamide, have with resistant bacteria.
been used in addition to prednisolone, but whether these drugs The prognosis for cats with FIP is generally grave. Almost all
improve outcome is unknown, and they have the potential to be cats with effusion at the time of diagnosis die within weeks. Very
toxic. A variety of immunomodulators and antiviral drugs have rarely, more prolonged survival times (1 to 2 years) have been
been tried, such as ribavirin and oral and parenteral human documented after glucocorticoid treatment (see Case Example).
204 SECTION 1  Viral Diseases

B
FIGURE 20-10  A, Gross necropsy findings in a 10-month-old male intact domes-
tic shorthair cat with effusive FIP. Approximately 1.2 liters of yellow fluid was present in
the abdomen, and there are abundant fibrin strains adherent to the visceral and parietal
peritoneal surfaces. B, Kidneys of the cat in Figure 20-8. Multiple, pale tan firm nodules
expand the renal cortices and protrude from the cortical surfaces. (Courtesy of the Univer-
sity of California, Davis Veterinary Anatomic Pathology service, D. Gasper and M. Jones.) B
FIGURE 20-11  A, Histopathology of the liver of a cat with FIP. There is pyogranulo-
matous hepatitis. Hematoxylin and eosin stain. B, Macrophages stain strongly positive for
The median survival time in one study of 37 cats was 9 days feline coronavirus antigen with immunohistochemistry. (Courtesy Dr. Patricia Pesavento,
University of California, Davis Veterinary Anatomic Pathology service.)
(range, 3 to 200 days).57 In another it was 21 days (range, 1
to 99 days) for 30 cats with effusive disease, 38 days (range,
1 to 171 days) for 12 cats with noneffusive disease, and 111
days (range, 7 to 477 days) for 9 cats with mixed effusive and TABLE 20-6
noneffusive disease.14 Hyperbilirubinemia, the presence of effu-
sion, and lymphopenia are negative prognostic factors20; in one Suggested Drug Dosages for Treatment of Feline
study, the hematocrit, lymphocyte count, and serum albumin, Infectious Peritonitis
potassium, sodium, and globulin concentrations decreased as
disease progressed, and total bilirubin concentration and serum Interval
liver enzyme activities increased.14 Euthanasia should be consid- Drug Dose (mg/kg) Route (hours)
ered for cats with severe illness that fail to respond to treatment
Prednisolone 1-2 PO 12-24
within a 3-day period.20
Chlorambucil* 2 mg/cat PO 48-72
Immunity and Vaccination *Monitor the CBC during treatment.

Although antibodies to the spike protein can neutralize virus,


antibodies are required for FIP to occur, because FIP is an
immune complex disease. Cell-mediated immunity is important known as antibody-dependent disease enhancement (ADDE).
for protection,6 but if immunity is incomplete, granulomatous Currently, an intranasal, temperature-sensitive mutant serotype
or pyogranulomatous disease results. II FIP virus vaccine is available on the market, but its use has
The development of vaccines for FIP has been complicated been controversial. The vaccine virus replicates in the lower
by the fact that stimulation of antibody production against temperatures found in the respiratory tract. It is licensed for
FCoV can accelerate the disease, should FIP develop after vacci- administration from 16 weeks of age, by which time most kit-
nation has been performed. Antibodies may bind to Fc receptors tens have already been exposed to FCoV. The vaccine does not
on macrophages and accelerate virus uptake in a phenomenon appear to cause ADDE,63-65 but its efficacy and ability to induce
CHAPTER 20  Feline Coronavirus Infection 205

immunity against heterologous strains is controversial. In a chronic shedders with serial fecal RT-PCR assays and removal
study of 138 cats that belonged to 15 different cat breeders, of kittens from the queen followed by isolation at 5 to 6 weeks
virtually all of which were seropositive, there was no difference of age (before maternal antibody has declined), have limita-
in prevalence of FIP in vaccinated versus placebo-treated cats.63 tions and are difficult to achieve properly in large catteries.20
A slight reduction in the prevalence of FIP occurred when the For example, cats that do not shed FCoV may still be infected
vaccine was used in cats that had not been exposed to FCoV with avirulent FCoV strains, and shedding may recommence at
before vaccination, but protection was not convincing based on a later date.22 Isolation of kittens may be useful if reexposure
the small numbers of cats that developed the disease in each is prevented until after they are 16 weeks of age, when their
group.6,65 immune system is more mature.6 In shelter situations, FIP may
be reduced when overcrowding and prolonged stays are mini-
Prevention mized, especially during kitten season.6 If possible, owners
that adopt cats from shelter environments should be provided
In households that contain only one or a few cats, young cats with a handout that provides basic information on the disease
that develop FIP likely become infected with FCoV before they (and other major infectious diseases of shelter cats such as ret-
are acquired. They may or may not have FIP at the time of rovirus infections, bartonellosis, and feline upper respiratory
acquisition. When a cat from a single-cat household dies with tract disease) and the ubiquitous nature of infection.
FIP, it is suggested that the owner wait at least 2 months before Further understanding of genetic factors that contribute to
a new cat is obtained, so that any virus in the environment FIP is required, because selective breeding may reduce the risk
becomes inactivated.20 Selection of a new cat from a different of the disease. In the meantime, the breeding of cats that pro-
genetic background than the previous cat should be considered, duce litters that succumb to FIP should be avoided. This is espe-
and if possible, the breeder should be informed if a purebred cially true for male cats, because a single male cat can have an
cat develops FIP. If a low number of other cats remain in the effect on far more kittens and litters than a single queen. It is
household, they may or may not continue to shed virus. These recommended that no more than six breeding animals be main-
cats often have a positive antibody titer, but this in no way tained if possible.6
predicts that they will develop FIP. Before a new cat is intro-
duced to a household that has a history of FIP, factors that Public Health Aspects
could reduce stress and overcrowding should be identified and
addressed. There is no evidence that humans can become infected with
The risk of transmission and disease can be reduced through FCoV. The closest human coronavirus relative is the severe
attention to hygiene, prevention of overcrowding, maintenance acute respiratory syndrome (SARS) coronavirus. Other corona-
of a larger ratio of adult to juvenile cats, and ensuring that cats viruses cause FIP-like disease in nonfelids such as ferrets and
are in stable groups of three or fewer per room. Cats should mice. If a coronavirus emerged that could cause similar clinical
have sufficient numbers of regularly cleaned litter trays located manifestations and outcomes in humans as FIPV can in cats, it
in a different area from where they are fed. Methods to control would represent a major threat to humans and would be the
FIP in cattery situations, such as identification and removal of subject of intense research.

CASE EXAMPLE after which treatment was discontinued. For the 3 weeks
that followed, the cat had been appetent and energetic, but
occasional soft feces had been noticed in the litter box. The
Signalment: “Ricky”, a 9-month-old male castrated domestic owners were concerned about the possibility of FIP.
shorthair from Sacramento, CA Ricky was obtained at 3 months of age from a rescue group,
History: Ricky was brought to a local veterinary clinic because who rescued him as an 8-week-old stray kitten. As a kitten
of increased thirst and urination. A serum chemistry panel he had multiple upper respiratory tract infections, but since
showed hyperglobulinemia (7.7 mg/dL), and urinalysis adoption he had been healthy and shared a household with
showed a specific gravity (SGr) of 1.025 with an inactive one other cat. He was an indoor cat that was sometimes
sediment; aerobic bacterial urine culture was negative. Three walked briefly outdoors. He was fed commercial dry and wet
days later, Ricky became inappetent and was returned to the cat food.
local veterinary clinic. Laboratory abnormalities included Physical Examination:
mature neutrophilia (11,904 cells/µL), lymphocytosis (5104 Body Weight: 4.2 kg
cells/µL), eosinophilia (1536 cells/µL), hyperglobulinemia General: Bright, alert and responsive, hydrated. T = 103°F
(6.9 mg/dL), and hypoalbuminemia (2.4 mg/dL). A feline (39.4°C), HR = 200 beats/min, eupneic.
coronavirus antibody titer was 1:400. Serology for Toxoplasma All Systems: No clinically significant abnormalities of any body
gondii was negative. Plain thoracic radiographs showed system were detected. Body condition score was 5/9.
a mild interstitial pattern. Abdominal ultrasound showed Laboratory Findings:
mesenteric lymphadenomegaly, and an aspirate of the CBC:
lymph nodes showed lymphoid reactivity. Treatment with HCT 27.3% (30%-50%)
cyproheptadine was initiated, and Ricky’s appetite recovered, MCV 42.1 fL (42-53 fL)
206 SECTION 1  Viral Diseases

MCHC 34.1 g/dL (30-33.5 g/dL) Treatment and Outcome: Biopsy of the enlarged mesenteric
WBC 24,950 cells/µL (4500-14,000 cells/µL) node was offered, but the owners declined. Ricky was treated
Neutrophils 17,141 cells/µL (2000-9000 cells/µL) with prednisolone (5 mg PO q12h for 7 days, followed by 5
Lymphocytes 6886 cells/µL (1000-7000 cells/µL) mg PO q24h thereafter), chlorambucil (2 mg PO every 3 days),
Monocytes 724 cells/µL (50-600 cells/µL) and pentoxifylline (50 mg PO q8h). Six weeks later, the cat
Eosinophils 200 cells/µL (150-1100 cells/µL) was well and CBC variables within reference ranges. Serum
Basophils 25 cells/µL (0-50 cells/µL) total protein concentration was 8.5 g/dL, with a globulin
Platelets 518,000/µL (180,000-500,000 platelets/µL). concentration of 4.5 g/dL. Abdominal ultrasound examination
Serum Chemistry Profile: showed persistent but mild mesenteric lymphadenomegaly
Sodium 147 mmol/L (151-158 mmol/L) (0.5 to 0.75 cm in diameter). A feline coronavirus titer was
Potassium 4.5 mmol/L (3.6-4.9 mmol/L) 1:25,600. Treatment with feline interferon-ω was commenced
Chloride 116 mmol/L (117-126 mmol/L) (4.5 million units SC once weekly). Ricky was seen again 3
Bicarbonate 18 mmol/L (15-21 mmol/L) months later, at which time he continued to be playful and
Phosphorus 6.4 mg/dL (3.2-6.3 mg/dL) appetent, with a stable body weight of 4.5 kg. A CBC showed
Calcium 9.5 mg/dL (9.0-10.9 mg/dl) mild anemia (HCT 29%), a neutrophil count of 4439 cells/µL,
BUN 22 mg/dL (18-33 mg/dL) and lymphopenia (468 cells/µL). A chemistry panel and
Creatinine 1.1 mg/dL (1.1-2.2 mg/dL) abdominal ultrasound showed no abnormalities. Chlorambucil
Glucose 83 mg/dL (63-118 mg/dL) and interferon-ω were discontinued. The next time the cat was
Total protein 10.7 g/dL (6.6-8.4 g/dL) reexamined was 12 months after the onset of illness, at which
Albumin 2.6 g/dL (2.2-4.6 g/dL) time he continued to be apparently healthy. CBC findings
Globulin 8.1 g/dL (2.8-5.4 g/dL) were unchanged, and the serum globulin concentration
ALT 31 U/L (27-101 U/L) was 4.7 g/dL. The prednisolone dose was decreased to 5 mg
AST 17 U/L (17-58 U/L) q48h and pentoxifylline treatment was discontinued. One
ALP 35 U/L (14-71 U/L) month later, albumin and globulin concentrations were 3.3
Gamma GT <3 U/L (0-4 U/L) and 5.1 g/dL, respectively, and the prednisolone dose was
Cholesterol 143 mg/dL (89-258 mg/dL) reduced to 2.5 mg q48h. At the next 1-month recheck, a CBC
Total bilirubin < 0.1 mg/dL (0-0.2 mg/dL). was unremarkable but globulin was 5.7 g/dL. Abdominal
Serum Protein Electrophoresis: A polyclonal gammopathy ultrasound showed mildly enlarged mesenteric lymph nodes,
with a mild decrease in albumin concentration and mild and the serum coronavirus antibody titer was 1:409,600. The
increases in the α2 and β1 fractions was present (see Figure prednisolone dose was increased to 5 mg PO q24h; 1 month
20-4). These changes were consistent with the acute-phase later, the serum globulin concentration was 4.9 g/dL. One and
inflammatory response. a half years after the onset of illness, Ricky was still apparently
Imaging Findings: Abdominal ultrasound: The spleen healthy according to the owner, but a midabdominal mass was
was moderately enlarged. There was diffuse mesenteric palpated on physical examination, and the serum globulin
lymphadenopathy (see Figure 20-8, B). concentration had increased again (5.6 g/dL). Abdominal
Mesenteric Lymph Node Aspirate Cytology: Intact nucleated ultrasound showed several moderately enlarged and
cells were composed of a heterogenous population of hypoechoic lymph nodes in the ileocolic region, the largest
lymphocytes, predominated by small, mature lymphocytes. of which was 0.9 cm in diameter. The surrounding mesentery
Lower numbers of intermediate and large reactive was focally hyperechoic. There was also focal hyperechoic
lymphocytes, moderate numbers of mildly degenerate retroperitoneal tissue surrounding the right kidney with scant
neutrophils, and scattered plasma cells and histiocytes retroperitoneal fluid. Attempts to obtain aspirates from the
were noted. Immunocytochemistry using two different lymph nodes were unsuccessful. The prednisolone dose was
monoclonal antibodies against FCoV was negative, but increased to 5mg PO q12h, and treatment with chlorambucil
macrophages were low in number. and pentoxifylline was reinstituted.
Microbiologic Testing: FeLV antigen and FIV antibody One week later, Ricky developed lethargy and inappetence. A
serology: negative CBC showed macrocytic anemia (HCT 24.3%, MCV 57.2 fL)
Serology (IFA) and blood culture for Bartonella clarridgeiae and and lymphopenia (782 cells/µL). A serum chemistry panel
Bartonella henselae: negative showed only hyperglobulinemia (5.8 g/dL). FeLV and FIV
Serology (IFA) for vector-borne diseases: negative for antibod- serology was repeated and was again negative, and the
ies to Ehrlichia canis, Neorickettsia risticii, Anaplasma spp., coronavirus antibody titer was 1:25,600. Treatment with
and Rickettsia spp. cyproheptadine was initiated and the chlorambucil and
PCR for FCoV (whole blood): negative pentoxifylline discontinued. However, inappetence contin-
PCR panel for other bloodborne pathogens (Anaplasma phago- ued, and persistent pyrexia (103.4° to 104°F), hematoche-
cytophilum, Anaplasma platys, Bartonella spp., E. canis, N. ris- zia, and tachypnea developed over the next few days. The
ticii, Mycoplasma haemofelis): negative hematocrit dropped to 16.5%, and hypokalemia, hyponatre-
Aerobic and anaerobic bacterial culture of mesenteric lymph mia, and hypochloremia were identified. Ricky was hospital-
node aspirate: negative ized and treated with 1 unit of packed RBC, IV crystalloids,
Serology for FCoV: positive at 1:102,400 and parenteral antimicrobial drugs. Thoracic radiographs
Diagnosis: A tentative diagnosis of FIP was made on the basis showed a severe, diffuse, patchy alveolar and nodular inter-
of Ricky’s background, the marked polyclonal gammopathy, stitial pattern with thickening of the bronchial walls and
and the strongly positive coronavirus titer. mild pleural effusion (see Figure 20-7). A tracheobronchial
CHAPTER 20  Feline Coronavirus Infection 207

lavage showed marked mixed, predominantly pyogranulo- Comments: The course of disease and survival time (587
matous inflammation with moderate epithelial hyperplasia days) in this cat was unusually prolonged for FIP, and on
and some degenerate neutrophils (see Figure 20-9). Immu- many occasions the diagnosis was questioned. However,
nocytochemistry with an anti-FCoV antibody was strongly the persistently increased FCoV antibody titer in a cat
positive in macrophages. Aerobic and anaerobic bacterial that lived with only one other cat raised suspicion for the
cultures of the wash specimen were negative. The cat subse- disease. Chronic, smoldering FIP may be more common
quently seizured and was euthanized. than recognized.6 The initial clinical signs in this cat were
Necropsy Findings: Necropsy showed moderate to severe, mild and may have been overlooked by some owners.
multifocal to coalescing pyogranulomatous capsulitis and Although the cat appeared to respond to prednisolone
serositis that involved the spleen, liver, kidney, intestines, treatment, it was not known whether the other medications
diaphragm, thoracic and abdominal walls, and pericardium. used had any effect. Ultimately, disease progressed, and a
There was also multifocal pyogranulomatous splenitis, diagnosis of FIP was confirmed with immunocytochemistry
hepatitis, nephritis, meningoencephalitis, and pneumonia on the tracheobronchial lavage specimen. Although
with necrosis. Straw-colored effusion was present in effusion developed, the amount was too low to permit
the abdominal cavity, thoracic cavity, and pleural space. collection of the fluid for analysis. The initial negative PCR
Immunohistochemistry was strongly positive for FCoV and immunocytochemistry results may have reflected the
antigen (see Figure 20-11). presence of low quantities of virus.

SUGGESTED READINGS 13. Norris JM, Bosward KL, White JD, et al. Clinicopathological find-
Addie D, Belak S, Boucraut-Baralon C, et al. Feline infectious peritoni- ings associated with feline infectious peritonitis in Sydney, Austra-
tis. ABCD guidelines on prevention and management. J Feline Med lia: 42 cases (1990-2002). Aust Vet J. 2005;83:666-673.
Surg. 2009;11:594-604. 14. Tsai HY, Chueh LL, Lin CN, et  al. Clinicopathological
Brown MA. Genetic determinants of pathogenesis by feline infectious findings and disease staging of feline infectious peritonitis: 51 cases
peritonitis virus. Vet Immunol Immunopathol. 2011;143:265-268. from 2003 to 2009 in Taiwan. J Feline Med Surg. 2011;13:74-80.
Pedersen NC. A review of feline infectious peritonitis virus infection: 15. Hartmann K. Feline infectious peritonitis. Vet Clin North Am
1963-2008. J Feline Med Surg. 2009;11:225-258. Small Anim Pract. 2005;35:39-79, vi.
16. Bell ET, Toribio JA, White JD, et al. Seroprevalence study of feline
REFERENCES coronavirus in owned and feral cats in Sydney, Australia. Aust Vet J.
2006;84:74-81.
1. Holzworth J. Some important disorders of cats. Cornell Vet.
17. Pedersen NC. Serologic studies of naturally occurring feline infec-
1963;53:157-160.
tious peritonitis. Am J Vet Res. 1976;37:1449-1453.
2. McArdle F, Bennett M, Gaskell RM, et al. Induction and enhance-
18. Addie DD, Jarrett O. A study of naturally occurring feline corona-
ment of feline infectious peritonitis by canine coronavirus.
virus infections in kittens. Vet Rec. 1992;130:133-137.
Am J Vet Res. 1992;53:1500-1506.
19. Addie DD, Toth S, Murray GD, et al. Risk of feline infectious peri-
3. Van Hamme E, Desmarets L, Dewerchin HL, et al. Intriguing inter-
tonitis in cats naturally infected with feline coronavirus. Am J Vet
play between feline infectious peritonitis virus and its receptors dur-
Res. 1995;56:429-434.
ing entry in primary feline monocytes. Virus Res. 2011;160:32-39.
20. Addie D, Belak S, Boucraut-Baralon C, et al. Feline infectious peri-
4. Tekes G, Hofmann-Lehmann R, Stallkamp I, et al. Genome orga-
tonitis. ABCD guidelines on prevention and management. J Feline
nization and reverse genetic analysis of a type I feline coronavirus.
Med Surg. 2009;11:594-604.
J Virol. 2008;82:1851-1859.
21. Gunn-Moore DA, Gruffydd-Jones TJ, Harbour DA. Detection of
5. Pedersen NC, Black JW, Boyle JF, et  al. Pathogenic differences
feline coronaviruses by culture and reverse transcriptase-polymerase
between various feline coronavirus isolates. Adv Exp Med Biol.
chain reaction of blood samples from healthy cats and cats with clini-
1984;173:365-380.
cal feline infectious peritonitis. Vet Microbiol. 1998;62:193-205.
6. Pedersen NC. A review of feline infectious peritonitis virus infec-
22. Kipar A, Meli ML, Baptiste KE, et  al. Sites of feline coronavirus
tion: 1963-2008. J Feline Med Surg. 2009;11:225-258.
persistence in healthy cats. J Gen Virol. 2010;91:1698-1707.
7. Hohdatsu T, Okada S, Ishizuka Y, et al. The prevalence of types
23. Chang HW, de Groot RJ, Egberink HF, et al. Feline infectious peri-
I and II feline coronavirus infections in cats. J Vet Med Sci.
tonitis: insights into feline coronavirus pathobiogenesis and epide-
1992;54:557-562.
miology based on genetic analysis of the viral 3c gene. J Gen Virol.
8. Heeney JL, Evermann JF, McKeirnan AJ, et  al. Prevalence and
2010;91:415-420.
implications of feline coronavirus infections of captive and free-
24. Rottier PJ, Nakamura K, Schellen P, et al. Acquisition of macro-
ranging cheetahs (Acinonyx jubatus). J Virol. 1990;64:1964-1972.
phage tropism during the pathogenesis of feline infectious peri-
9. Rohrbach BW, Legendre AM, Baldwin CA, et al. Epidemiology of
tonitis is determined by mutations in the feline coronavirus spike
feline infectious peritonitis among cats examined at veterinary med-
protein. J Virol. 2005;79:14122-14130.
ical teaching hospitals. J Am Vet Med Assoc. 2001;218:1111-1115.
25. Chang HW, Egberink HF, Halpin R, et  al. Spike protein fusion
10. Pesteanu-Somogyi LD, Radzai C, Pressler BM. Prevalence of feline
peptide and feline coronavirus virulence. Emerg Infect Dis.
infectious peritonitis in specific cat breeds. J Feline Med Surg.
2012;18(7):1089-1095.
2006;8:1-5.
26. Brown MA, Troyer JL, Pecon-Slattery J, et al. Genetics and patho-
11. Worthing KI, Wigney DI, Dhand NK, et al. Risk factors for feline
genesis of feline infectious peritonitis virus. Emerg Infect Dis.
infectious peritonitis in Australia. J Fel Med Surg. 2012;14:405-412.
2009;15:1445-1452.
12. Foley JE, Poland A, Carlson J, et  al. Risk factors for feline
27. Vennema H, Poland A, Foley J, et al. Feline infectious peritonitis
infectious peritonitis among cats in multiple-cat environments
viruses arise by mutation from endemic feline enteric coronavi-
with endemic feline enteric coronavirus. J Am Vet Med Assoc.
ruses. Virology. 1998;243:150-157.
1997;210:1313-1318.
208 SECTION 1  Viral Diseases

28. Pedersen NC, Liu H, Dodd KA, et al. Significance of coronavirus 46. Duthie S, Eckersall PD, Addie DD, et al. Value of alpha 1-acid gly-
mutants in feces and diseased tissues of cats suffering from feline coprotein in the diagnosis of feline infectious peritonitis. Vet Rec.
infectious peritonitis. Viruses. 2009;1:166-184. 1997;141:299-303.
29. Takano T, Tomiyama Y, Katoh Y, et al. Mutation of neutralizing/ 47. Giori L, Giordano A, Giudice C, et  al. Performances of different
antibody-dependent enhancing epitope on spike protein and 7b diagnostic tests for feline infectious peritonitis in challenging clini-
gene of feline infectious peritonitis virus: influences of viral replica- cal cases. J Small Anim Pract. 2011;52:152-157.
tion in monocytes/macrophages and virulence in cats. Virus Res. 48. Savary KC, Sellon RK, Law JM. Chylous abdominal effusion in
2011;156:72-80. a cat with feline infectious peritonitis. J Am Anim Hosp Assoc.
30. Brown MA. Genetic determinants of pathogenesis by feline 2001;37:35-40.
infectious peritonitis virus. Vet Immunol Immunopathol. 49. Shelly SM, Scarlett-Kranz J, Blue JT. Protein electrophoresis on
2011;143:265-268. effusions from cats as a diagnostic test for feline infectious perito-
31. Addie DD, Schaap IA, Nicolson L, et al. Persistence and transmis- nitis. J Am Anim Hosp Assoc. 1988;24:495-500.
sion of natural type I feline coronavirus infection. J Gen Virol. 50. Fischer Y, Sauter-Louis C, Hartmann K. Diagnostic accuracy
2003;84:2735-2744. of the Rivalta test for feline infectious peritonitis. Vet Clin Pathol.
32. Addie DD, Jarrett O. Use of a reverse-transcriptase polymerase 2012;41:558-567.
chain reaction for monitoring the shedding of feline coronavirus by 51. Boettcher IC, Steinberg T, Matiasek K, et al. Use of anti-corona-
healthy cats. Vet Rec. 2001;148:649-653. virus antibody testing of cerebrospinal fluid for diagnosis of feline
33. Pedersen NC, Allen CE, Lyons LA. Pathogenesis of feline enteric infectious peritonitis involving the central nervous system in cats.
coronavirus infection. J Feline Med Surg. 2008;10:529-541. J Am Vet Med Assoc. 2007;230:199-205.
34. Regan AD, Ousterout DG, Whittaker GR. Feline lectin activity is 52. Foley JE, Lapointe JM, Koblik P, et  al. Diagnostic features of
critical for the cellular entry of feline infectious peritonitis virus. clinical neurologic feline infectious peritonitis. J Vet Intern Med.
J Virol. 2010;84:7917-7921. 1998;12:415-423.
35. Takano T, Ohyama T, Kokumoto A, et  al. Vascular endothe- 53. Lewis KM, O’Brien RT. Abdominal ultrasonographic findings
lial growth factor (VEGF), produced by feline infectious perito- associated with feline infectious peritonitis: a retrospective review
nitis (FIP) virus-infected monocytes and macrophages, induces of 16 cases. J Am Anim Hosp Assoc. 2010;46:152-160.
vascular permeability and effusion in cats with FIP. Virus Res. 54. Pratelli A. Comparison of serologic techniques for the detec-
2011;158:161-168. tion of antibodies against feline coronaviruses. J Vet Diagn Invest.
36. Kiss I, Poland AM, Pedersen NC. Disease outcome and cytokine 2008;20:45-50.
responses in cats immunized with an avirulent feline infectious 55. Tammer R, Evensen O, Lutz H, et  al. Immunohistological dem-
peritonitis virus (FIPV)-UCD1 and challenge-exposed with virulent onstration of feline infectious peritonitis virus antigen in paraf-
FIPV-UCD8. J Feline Med Surg. 2004;6:89-97. fin-embedded tissues using feline ascites or murine monoclonal
37. Takano T, Azuma N, Satoh M, et al. Neutrophil survival factors antibodies. Vet Immunol Immunopathol. 1995;49:177-182.
(TNF-alpha, GM-CSF, and G-CSF) produced by macrophages 56. Ishida T, Shibanai A, Tanaka S, et  al. Use of recombinant feline
in cats infected with feline infectious peritonitis virus contrib- interferon and glucocorticoid in the treatment of feline infectious peri-
ute to the pathogenesis of granulomatous lesions. Arch Virol. tonitis. J Feline Med Surg. 2004;6:107-109.
2009;154:775-781. 57. Ritz S, Egberink H, Hartmann K. Effect of feline interferon-omega
38. de Groot-Mijnes JD, van Dun JM, van der Most RG, et  al. on the survival time and quality of life of cats with feline infectious
Natural history of a recurrent feline coronavirus infection and peritonitis. J Vet Intern Med. 2007;21:1193-1197.
the role of cellular immunity in survival and disease. J Virol. 58. Legendre AM, Bartges JW. Effect of Polyprenyl Immunostimulant
2005;79:1036-1044. on the survival times of three cats with the dry form of feline infectious
39. Davidson BJ, Paling AC, Lahmers SL, et al. Disease association and peritonitis. J Feline Med Surg. 2009;11:624-626.
clinical assessment of feline pericardial effusion. J Am Anim Hosp 59. Watari T, Kaneshima T, Tsujimoto H, et al. Effect of thromboxane
Assoc. 2008;44:5-9. synthetase inhibitor on feline infectious peritonitis in cats. J Vet Med
40. Bradshaw JM, Pearson GR, Gruffydd-Jones TJ. A retrospective Sci. 1998;60:657-659.
study of 286 cases of neurological disorders of the cat. J Comp 60. Hsieh LE, Lin CN, Su BL, et  al. Synergistic antiviral effect of
Pathol. 2004;131:112-120. Galanthus nivalis agglutinin and nelfinavir against feline coronavirus.
41. Hayashi T, Ishida T, Fujiwara K. Glomerulonephritis asso- Antiviral Res. 2010;88:25-30.
ciated with feline infectious peritonitis. Nihon Juigaku Zasshi. 61. McDonagh P, Sheehy PA, Norris JM. In vitro inhibition of feline
1982;44:909-916. coronavirus replication by small interfering RNAs. Vet Microbiol.
42. Taylor SS, Tappin SW, Dodkin SJ, et al. Serum protein electropho- 2011;150:220-229.
resis in 155 cats. J Feline Med Surg. 2010;12:643-653. 62. Tanaka Y, Sato Y, Osawa S, et al. Suppression of feline coronavirus
43. Sparkes AH, Gruffydd-Jones TJ, Harbour DA. Feline infec- replication in vitro by cyclosporin A. Vet Res. 2012;41-43.
tious peritonitis: a review of clinicopathological changes in 65 63. Fehr D, Holznagel E, Bolla S, et al. Placebo-controlled evaluation of a
cases, and a critical assessment of their diagnostic value. Vet Rec. modified live virus vaccine against feline infectious peritonitis: safety
1991;129:209-212. and efficacy under field conditions. Vaccine. 1997;15:1101-1109.
44. Hartmann K, Binder C, Hirschberger J, et al. Comparison of differ- 64. Postorino Reeves NC, Pollock RV, Thurber ET. Long-term follow-
ent tests to diagnose feline infectious peritonitis. J Vet Intern Med. up study of cats vaccinated with a temperature-sensitive feline infectious
2003;17:781-790. peritonitis vaccine. Cornell Vet. 1992:82.
45. Jeffery U, Dietz K, Hostetter S. Positive predictive value of 65. Postorino Reeves NC. Vaccination against naturally-occurring FIP
albumin:globulin ratio for feline infectious peritonitis in a mid-west- in a single large cat shelter. Feline Pract. 1995;23:81-82.
ern referral hospital population. J Fel Med Surg. 2012;14:903-905.
CHAPTER 21

Feline Immunodeficiency Virus Infection


Jane E. Sykes

which is expressed on feline CD4+ T cells, B cells, and activated


Overview of Feline Immunodeficiency Virus macrophages,6,7 and the secondary receptor CXCR4, which
Infection is normally a chemokine receptor. The viral envelope fuses
First Described: California, 1986 (Pedersen)1; serologic evi- with the cell membrane, and the capsid enters the cytoplasm,
dence of infection dates back to the 1960s2,3 where reverse transcription occurs and a double-stranded DNA
Cause: Feline immunodeficiency virus (FIV) (family Retroviri- (dsDNA) copy of the retroviral genome is made. Additional
dae, subfamily Orthoretrovirinae, genus Lentivirus) sequences, known as long terminal repeats (LTRs), are added
to either end of the viral genome. The virus then passes into the
Affected Hosts: Domestic and wild cats; hyenas
nucleus where the dsDNA copy integrates into the host genome;
Geographic Distribution: Worldwide at this point the integrated dsDNA becomes a provirus. Tran-
Mode of Transmission: Biting, and to a lesser extent transpla- scription of this DNA, which is controlled by the LTRs, leads
cental, through milk, possibly venereal, blood transfusion to synthesis of new virion components, and virus assembly and
budding occur at the cell surface (Figure 21-2). Depending on
Major Clinical Signs: Lethargy, fever, pallor, stomatitis, diar- the cellular environment, proviral DNA may either be latent,
rhea, muscle atrophy, neurologic signs, signs of under- whereby transcription does not occur, or be transcriptionally
lying neoplastic or immune-mediated disorders or active. Latency is one mechanism by which retroviruses can
opportunistic infections evade the host immune system. Because the reverse transcrip-
Differential Diagnoses: FeLV infection, bartonellosis, other tase enzyme is prone to error, the mutation rate of retroviruses
causes of stomatitis such as feline calicivirus infection, is high, and a diversity of viral variants continuously emerges
immune-mediated disease, other chronic inflammatory from infected hosts. Integration of retroviral DNA into host cell
and neoplastic diseases of cats that occur in the absence DNA can disrupt genes that are responsible for cell growth and
of detectable infection with FIV. differentiation (proto-oncogenes). Alternatively, cellular onco-
Human Health Significance: FIV does not infect humans. genes captured and carried by retroviruses develop mutations
during retroviral replication, and are then re-inserted into host
cell DNA. The end result is abnormal cell growth and differen-
tiation, which results in tumor formation by some retroviruses.
Etiology and Epidemiology Based on sequence diversity of the env gene, there are six
different subtypes of FIV, A through F. Subtypes A and B are
Feline immunodeficiency virus (FIV) is an enveloped, RNA distributed most widely, followed by subtype C8-10—although a
virus that belongs to the Lentivirus genus of the Retroviridae. recent study from the United States showed an equal distribu-
It infects domestic and wild cats worldwide, as well as hyenas.4 tion of subtypes A, B, and C (Table 21-1).11-31 In addition, a
Like HIV, FIV establishes a chronic, persistent infection that, in number of recombinant subtypes have been recognized, such
some cats, ultimately culminates in immunodeficiency. Because as A/B, A/C, B/D, B/E, and A/B/C recombinants, and additional
of its similarities to HIV, FIV infection in cats has been used as subtypes also likely exist.9,11 The heterogeneity of the virus
a research model for HIV infection and acquired immunodefi- complicates the design of molecular diagnostic tests and vac-
ciency syndrome (AIDS),5 and relative to other viral infections cines for FIV. Whether differences in the clinical manifestations
of companion animals, much is known about its pathogenesis. of disease relate to infection by different subtypes requires fur-
Knowledge of retroviral structure and replication is required ther study.9,10,31
in order to understand diagnostic, treatment, and prevention Retroviruses survive only minutes outside the host and
strategies that target these viruses. Like other retroviruses, FIV are very susceptible to disinfection. FIV is shed in high con-
has a three-layered structure that is composed of an inner- centrations in saliva, and the major mode of transmission is
most genome-nucleocapsid complex with helical symmetry, an through bites. Transplacental transmission, transmission dur-
icosahedral capsid, and an envelope with glycoprotein spikes ing parturition, and through milk have been documented
(­Figure 21-1). The FIV genome contains three major genes: gag, experimentally, but these modes appear to be uncommon in
which encodes the virion core proteins (capsid [p24], nucleo- naturally infected cats, and kittens infected by this route may
capsid, and matrix); pol, which encodes the reverse transcrip- not sustain productive infection.32,33 Venereal transmission has
tase, protease and integrase enzymes; and env, which encodes not been demonstrated, but FIV can be recovered from semen,
surface (gp120) and transmembrane virion (gp41) envelope and experimental inoculation of FIV into the vaginas of queens
glycoproteins. Several other accessory and regulatory genes are results in transmission.34,35 Transmission also has the potential
also present. FIV invades cells via the primary receptor CD134, to occur through blood donation.

209
210 SECTION 1  Viral Diseases

Lipid bilayer

p17
matrix gp41
RNA gp120

Reverse
transcriptase
Protease
Integrase

p24 capsid

Chemokine
CD134 receptor

T lymphocyte
surface

FIGURE 21-1  Structure of FIV. An FIV virion is shown adjacent to the surface of a CD4+ T cell. Lentiviruses contain two identical strands of RNA (the viral genome) and associated
enzymes, which include reverse transcriptase, integrase, and protease, packaged into a core composed of the p24 capsid protein with a surrounding p17 protein matrix, all enclosed by a
phospholipid membrane envelope that is derived from the host cell. Viral encoded membrane proteins (gp41 and gp120) are bound to the envelope. CD134 and CXCR4 (chemokine) recep-
tors on the host cell surface function as the receptors for FIV.

Seropositivity to FIV (which is equivalent to infection because dendritic cells, microglia, and astrocytes. The subsequent effects
of viral persistence) is consistently associated with a history of of the virus on the immune system are complex, incompletely
bite wounds, older age, male sex, illness, and outdoor access understood, and seem to result in both immune suppression and
(Table 21-2).12,36-41 Being of mixed breed has also been a risk immune activation.
factor in some studies. The mean age at diagnosis is around 6 Three phases of disease have been delineated, acute (pri-
to 8 years, and 80% to 90% of cats are more than 2 years of mary), subclinical, and terminal, although not all phases are
age. Male cats are up to 4.7 times more likely to be seroposi- recognized in many naturally infected cats. After inoculation,
tive than female cats.40 Indoor housing decreases transmission the virus replicates in lymphoid tissues, and high concentrations
but does not eliminate it.42 Worldwide, the seroprevalence of of virus are present in blood 2 weeks after infection. A peak
FIV in domestic pet cats ranges from around 1% to 12%.9,40,41 of viremia occurs 8 to 12 weeks after infection (Figure 21-3).
A study that included more than 18,000 North American cats There is a decline in CD4+ and CD8+ T cells in peripheral
estimated the overall prevalence of FeLV and FIV infections as blood. This may be associated with transient illness, which lasts
around 2.3% and 2.5%, respectively.5 Higher prevalences are 3 to 6 months and is often unrecognized by cat owners. Some
found in feral and free-ranging cats and sick cats. In the North cats show lethargy, fever, anorexia, diarrhea, stomatitis, weight
American study, the prevalence of FIV infection in sick, feral loss, and/or lymphadenopathy during the acute phase. Lymph-
cats was 18.2%, whereas that in healthy indoor cats was only adenopathy results from lymphoid hyperplasia, and can persist
0.7%. A study from France showed a seroprevalence of 21% in for weeks to months. Neutropenia can also occur,44 possibly
unowned cats, compared with 10% in owned cats.43 In Japan, as a result of neutrophil apoptosis. CD4+/CD25+ T regulator
the overall seroprevalence is very high (23% in one study), and (Treg) cells are infected and activated during the acute phase.
as many as one-third of male cats test positive.12 Occasionally, These cells then inhibit the proliferation of activated CD4+ and
co-infections with FeLV occur, and infection with one retrovirus CD8+ T cells, and cause them to undergo apoptosis. This may
is a risk factor for infection with the other.36,37 contribute to persistence of FIV and further immunosuppres-
sion.45-47 Altered dendritic cell function may also occur.48,49 In
Clinical Features general, impaired T cell function in acute infection is thought to
result from cytokine dysregulation, immunologic anergy (failure
Signs and Their Pathogenesis to respond to specific antigens), and increased apoptosis.45 Nev-
The main cellular target for FIV is the CD4+ T cell. How- ertheless, most cats survive this phase because of a rebound in
ever, FIV also infects CD8+ T cells, B cells, macrophages, and CD8+ T cell numbers and a strong humoral immune response.
CHAPTER 21  Feline Immunodeficiency Virus Infection 211

Virion binding
to CD134 and
chemokine
receptor New FIV
Fusion of FIV virion
membrane with host
FIV virion cell membrane; entry
of viral genome
into cytoplasm
Plasma
membrane Cytokine

Cytokine
receptor FIV
gp120/ Budding
Chemokine gp41
CD134 and release of
receptor
mature virion
FIV RNA
genome

Reverse
transcriptase–
mediated synthesis
of proviral DNA
Cytokine activation
of cell; transcription
Integration of of FIV genome;
provirus into transport of viral FIV core
host cell RNAs to cytoplasm structure
genome

Synthesis of
FIV proteins;
FIV DNA assembly of virion
provirus core structure
FIV RNA
transcript
Nucleus

FIGURE 21-2  The life cycle of FIV. The sequential steps in FIV reproduction are shown, from initial infection of a host cell to release of a new virus particle. An infected cell produces
many virions, each capable of infecting nearby cells, with subsequent spread of the infection.

hyperactivation. Some studies also describe a sustained increase


TABLE 21-1 in CD8+ T cell numbers. Although activated, paradoxically, T
cells have a reduced ability to respond to antigenic stimulation.
Distribution of FIV Subtypes Worldwide
Altered lymphocyte expression of cell surface molecules (includ-
A Australia, New Zealand, United States (especially ing CD4 and cytokine receptors, and MHC II antigens), and
western United States), South Africa, northwestern continued alteration of dendritic cell and neutrophil function
Europe, Japan, United Kingdom11-19 also contribute to immunosuppression. Dysregulation of cyto-
kine production occurs. For example, cats that are chronically
B Central and eastern United States, Caribbean, central
infected with FIV fail to produce Il-2, Il-6, and Il-12 in response
and western Europe, Brazil, eastern Japan11,14,20-25
to Toxoplasma gondii infection and instead produce elevated
C United States, Canada, New Zealand, southeast levels of the antiinflammatory cytokine Il-10.45,50 There is a
Asia11,12,26-29 slow influx of immune cells into the brain, with gradual pro-
D Japan, Vietnam, rarely United States21,27,31 gression of central nervous system (CNS) disease.51 The rate of
E Argentina10,30 progression of the subclinical phase depends on factors such as
the virus strain, co-infections with other agents that activate
F United States10,31 virus transcription, and host immunity.
In some cats, these changes ultimately lead to the terminal
phase, which is characterized by clinical signs of opportunistic
infections, neoplastic disease, myelosuppression, and neurologic
In the subclinical (or asymptomatic) phase, CD4+ T cell disease. This is the phase most commonly recognized in natu-
numbers rebound, and the plasma virus load declines to very rally infected cats (Table 21-3). However, many infected cats
low levels. Cats remain subclinically infected, often for years never develop FIV-related clinical signs, even when CD4+ T cell
or even for life. This is not latent infection, because virus pro- counts are low, and instead die from other causes. The terminal
duction continues at low levels; a slow, progressive decline in phase of FIV infection is commonly associated with moderate
CD4+ T cell numbers; reduction in the CD4+:CD8+ T cell ratio; to severe periodontal disease, lymphoplasmacytic stomatitis
and in some cats, hyperglobulinemia, which results from B cell (Figure 21-4), gingivitis, and feline odontoclastic resorptive
212 SECTION 1  Viral Diseases

lesions,52 which may result from opportunistic bacterial and


TABLE 21-2 viral infections. Other opportunistic infections include chronic
Risk Factors for FIV Infection in Cats Seen at the UC Davis VMTH bacterial skin and ear infections, persistent viral upper respira-
tory tract infections, dermatophytosis, mycobacterial infections,
Number Number fungal infections such as cryptococcosis and sporotrichosis,
of FIV+ of Control Odds hemoplasmosis, toxoplasmosis, and/or parasitic infections such
Variable Cats* Cats† Ratio P-value‡ as demodecosis and severe flea burdens. With the exception of
stomatitis, the relationship between many of these opportunistic
Sex infections and FIV infection is somewhat unclear, because the
Male neutered 100 117 — prevalence of many of these infections in cats with FIV infection
Male intact 10 5 2.729 0.08 is similar to that in cats without FIV infection. However, infec-
Female/female 17 88 0.24Åò <0.001 tions are often more severe and less responsive to treatment than
neutered the same infections in immunocompetent cats.
Breed The development of neoplasia in FIV-infected cats is thought
Mixed 116 170 — to result primarily from immune suppression, although there is
Purebred 11 40 0.37 0.02 serologic evidence that cats may be infected with a gammaher-
Environment pesvirus similar to Epstein-Barr virus that might be reactivated
Indoor 15 57 — in cats infected with FIV.53 Lymphomas are the most commonly
Outdoor 85 107 2.58 0.01 reported FIV-associated tumor, especially B cell lymphomas, but
also T cell and non-B, non-T cell lymphomas.54 FIV-infected
Stray history
cats are 5 times more likely to develop lymphoma than cats not
No 101 195 —
infected with FIV and are more likely to develop it an earlier age.
Yes 26 15 3.73 <0.001
Leukemia and squamous cell carcinomas (SCCs) are also com-
Cats in household mon tumors in cats with FIV, although the association with SCC
<3 34 70 — may be confounded by outdoor exposure. Other tumors include
≥3 39 61 1.25 0.53 mast cell tumors, fibrosarcomas, meningiomas, and metastatic
carcinomas; some infected cats develop more than one type of
Modified from Trott KA, Kass PH, Sparger EE, et  al. A clinical case
neoplasia. Rarely, lymphoma may develop as a result of viral inte-
control study: clinical presentation of FIV-positive cats. University of
California, Davis, STARS in Science Day. 2007; abstr. gration into the genome and disruption of proto-oncogenes.54,55
*All cats were >6 months of age and none had a history of FIV vaccination. Immune dysregulation and increased circulating immune
†All control cats were negative for FIV antibody at their visit to the complexes in the terminal phase can lead to immune-mediated
University of California, Davis. disorders, such as immune-mediated glomerulonephritis and
‡P-values <0.05 were significant. uveitis.56 Myelodysplasia develops in some cats,57,58 which
ÅòIn other words, female cats were four times less likely to be FIV+ than
may be manifested by clinical signs of lethargy, inappetence,
male neutered cats. pallor, or evidence of bleeding tendencies such as petechial
hemorrhages.

1200
-Possible acute HIV syndrome Death
-Wide dissemination of virus
1000 -Seeding of lymphoid organs
Opportunistic
infections 1:256
800
Plasma viremia titer
CD4+ T cells/mm3

Clinical latency

600 Constitutional 1:64


symptoms
400 1:16

200 1:4

0 0
0 6 12 1 5 10
Weeks Years
FIGURE 21-3  Changes in virus load and CD4+ T Cell numbers over the course of infection with HIV. A similar clinical course of infection occurs for FIV infection. Viremia is detected early
after infection and may be accompanied by systemic signs (acute phase). Plasma viremia then falls to very low levels and remains this way for many years (subclinical phase). Although this period
is referred to here as “clinical latency,” the virus itself is not latent during this time, because there is ongoing production of virus and a steady decline in CD4+ T cell counts. In the terminal phase
of infection, signs of immunodeficiency develop and plasma viremia increases. Some infected cats never reach this phase. Antibody production declines, and antibody tests may be negative in
cats with advanced terminal-phase disease, but PCR assays are more likely to be positive.
CHAPTER 21  Feline Immunodeficiency Virus Infection 213

TABLE 21-3
Disease Diagnoses in 127 Cats with FIV Infection and 210 Age-
Matched Control Cats Seen over the Same Time Period at the UC
Davis VMTH*
Cats Infected
with FIV Controls
Disease (n = 127)† (n = 210)‡ P-value§
Intraocular 16 17 0.18
­inflammation
Stomatitis 21 11 < 0.001
Cardiomyopathy 21 17 0.02
Upper respiratory 13 9 0.03 A
tract disease
Neurologic signs 16 24 0.75
Chronic kidney 23 40 0.83
disease
Diabetes mellitus 9 18 0.63
Hyperthyroidism 11 14 0.49
Lymphoma 23 20 0.02
Squamous cell 12 9 0.057
­carcinoma (SCC)
Fibrosarcoma 2 9 0.17
Carcinomas other 4 12 0.28
than SCC

Modified from Trott KA, Kass PH, Sparger EE, et  al. A clinical case
control study: clinical presentation of FIV-positive cats. University of
California, Davis, STARS in Science Day. 2007; abstr.
*Results should be interpreted with caution because the data were retro-
spectively collected. Some cats may have had undiagnosed disease, such
as subclinical chronic kidney or cardiac disease. The pathogenesis of a
disease process in FIV-positive cats might also differ from that found in B
FIV-negative cats.
†All cats were >6 months of age and none had a history of FIV vaccina- FIGURE 21-4  severe lymphoplasmacytic stomatitis in a cat infected with feline
tion.
immunodeficiency virus. A, There is marked hyperemia, ulceration, and proliferative
‡All control cats were negative for FIV antibody at their visit to the lesions in the palatoglossal folds. (From Bonello D, Roy CG, Verstraete FJM. Non-neoplastic
University of California, Davis.
proliferative oral lesions. In: Verstraete FJM, Lommer MJ, eds. Oral and Maxillofacial Sur-
§As determined by chi-square (univariate) analysis. P-values <0.05 were gery in Dogs and Cats. Philadelphia, PA: Saunders; 2011.) B, Histopathology of a biopsy
significant.
from a cat with severe stomatitis associated with FIV infection.

degenerative changes in other organs, such as the myocardium


The end result of infection by neurovirulent strains of FIV and the kidneys, is not well understood, because the prevalence
can be progressive behavioral changes such as increased aggres- of cardiomyopathy and interstitial nephritis in geriatric cats not
sion and cognitive disturbances, tremors, sleep disturbances, infected with FIV is high (see Table 21-3).
anisocoria, delayed reflexes, abnormal cranial nerve function, When transplacental transmission occurs in FIV-infected
urinary and fecal incontinence, and seizures. Decreased nerve queens, only some kittens in a litter may be infected. In the first
conduction velocities and abnormal electroencephalograms and year of infection, the overall rate of such transmission is around
brainstem evoked potentials have been documented.59 The virus 70%.62 Rates of mother-to-kitten transmission are highest in
does not infect neurons, but neuronal cell death occurs through queens that have a CD4+ count less than 200 cells/µL, those
multiple incompletely defined mechanisms.51 with signs of immunodeficiency, and those infected within the
Inflammatory lesions can develop in a variety of organs in past 15 months.63 In utero transmission can lead to arrested
cats infected with FIV. An inflammatory myopathy has been fetal development, abortion, stillbirth, low birth weights, and
described,60 and terminally, severe muscle atrophy can occur. the birth of T cell deficient kittens.62,64
Infection is associated with gut inflammation and intestinal epi-
thelial cell damage (also known as AIDS enteropathy), which may Physical Examination Findings
result in chronic diarrhea and failure to gain weight.61 The extent Many cats infected with FIV have no signs of illness, or clini-
to which chronic FIV infection contributes to inflammatory or cal signs that are unrelated to FIV infection. Cats in the acute
214 SECTION 1  Viral Diseases

TABLE 21-4
Diagnostic Assays Available for Feline Immunodeficiency Virus Infection
Assay Specimen Type Target Performance
Serology (ELISA, Blood, serum Antibody to FIV Positive test results in cats >6 months of age that have not been
Western blotting, vaccinated for FIV equal infection, but confirmation is recom-
IFA) mended in healthy cats using a second test from a different
manufacturer or Western blot. False positives occur in cats <6
months of age (maternal antibody) and cats vaccinated with the
FIV vaccine. False negatives can occur in the first 2 months of in-
fection or with advanced disease and severe immunosuppression.
PCR Blood FIV proviral DNA Sensitivity and specificity vary between laboratories. Even well-
or viral RNA designed assays may be insensitive because of viral variability
and low-level viremia. PCR assays are the only assay that can
definitively diagnose infection in cats vaccinated for FIV. Never
use without performing serology.
Virus ­isolation Blood FIV Difficult, not widely available. Used primarily as a research tool.

IFA, immunofluorescent antibody.

phase of FIV infection may be febrile or show generalized


peripheral lymphadenopathy. Behavioral abnormalities such
BOX 21-1
as obtundation, aggression, and cognitive impairment may be
Indications for Serologic Testing for FIV and FeLV
evident in cats with terminal neurologic disease; other neuro-
logic abnormalities include ataxia, anisocoria, and abnormal
segmental reflexes. Ocular abnormalities may be present as • Sick cats, even if tested negative in the past
a result of the FIV infection itself, opportunistic infections, • All newly acquired cats or kittens (2 tests, at least 60
or lymphoma, and include anterior uveitis, hyphema, pars days apart)
planitis, chorioretinitis, and/or glaucoma.65 Other common • After exposure to a retrovirus-infected cat or a cat
physical examination findings in cats with terminal disease are with unknown status, and especially after a bite wound
periodontal disease and chronic stomatitis, signs of chronic (2 tests, at least 60 days apart)
upper respiratory tract infection, otitis externa, pyoderma, • Cats that live in a household with other retrovirus-
cutaneous abscesses, and a diverse and variable spectrum of infected cats (annual retesting unless isolated)
disease manifestations that relate to other underlying opportu- • Before initial vaccination with FeLV or FIV vaccines
nistic infections or neoplasia. • Before use as a blood donor (in conjunction with real-
time PCR)
• On entry to a shelter or before adoption (2 tests, at
Diagnosis least 60 days apart); if financial resources are not avail-
For many cats, infection with FIV is diagnosed during screening able for this, cats should be held singly and postadop-
efforts. Screening for infection should be performed with tests tion testing (2 tests, at least 60 days apart) recommend-
that detect antibody against FIV, because these assays have the ed before mingling with other cats occurs. The status of
highest overall sensitivity and are rapid and widely available the other cats in the household should be known before
(Table 21-4). It has been recommended that the retrovirus status new cats are introduced.
of all cats be known regardless of the presence of absence of ill- • For group-housed cats, before introduction (2 tests, at
ness.66 Indications for testing as recommended by the American least 60 days apart if possible) and on an annual basis
Association of Feline Practitioners (AAFP) are shown in Box • Testing is considered optional for feral cat trap-neuter-
21-1. In practice, compliance with retrovirus testing is low. In return programs
a study that evaluated 967 cats with bite wounds or cutaneous
abscesses that presented to veterinary practitioners from 134 Modified from Levy J, Crawford C, Hartmann K, et al. 2008 Ameri-
can Association of Feline Practitioners’ feline retrovirus management
practices in 30 states, the combined FeLV-FIV status of only 96
guidelines. J Feline Med Surg 2008;10:300-316.
(9.9%) of the cats was known.38 In addition, despite the avail-
ability of a financial incentive for retesting, only 64 of 478 cat
owners returned their cats for retesting after treatment.
When positive test results occur in sick cats, the role that FIV availability of test methodologies in the future, clinical assess-
plays as a cause of the signs may be unclear, although it is reason- ment of CD4+ T cell counts, the CD4+/CD8+ ratio, and plasma
able to assume that FIV may be playing a role in cats with severe viral loads may facilitate interpretation of the relationship
stomatitis, unusual infections with intracellular pathogens such between disease manifestations and infection and provide prog-
as mycobacteria, and lymphoma. With refinement and improved nostic information, as in human patients infected with HIV.67
CHAPTER 21  Feline Immunodeficiency Virus Infection 215

Laboratory Abnormalities used to screen healthy cats for infection, confirmation of posi-
Complete Blood Count tive results is recommended because of the low prevalence of
Common abnormalities on the CBC in cats infected with FIV infection in this population of cats and the higher possibility
include mild anemia, lymphopenia, and neutropenia. Occasion- that false-positive test results may occur. Confirmation can be
ally severe anemia, thrombocytopenia, thrombocytosis, mono- done using an assay from a different manufacturer or Western
cytopenia, or leukocytosis occur. In one large European study, blotting. However, Western blotting is technically demanding,
neutrophil counts of FIV-infected cats were lower than those of subject to interoperator variability in interpretation, and it may
control cats, and lymphocyte counts were higher than those of be less sensitive than ELISA assays when performed by some
control cats.68 Leukopenia and neutropenia were more likely to laboratories.72
be present in FIV-infected cats. Positive ELISA assay results in the absence of FIV infec-
tion can occur in cats that have been vaccinated for FIV, or
Serum Biochemical Tests kittens less than 6 months of age that possess maternal anti-
The most common and significant abnormality on the chemis- body (because of infection or vaccination of the queen). No
try panel in cats infected with FIV is hyperproteinemia, which currently available serologic test, including Western immu-
results from increased γ-globulin concentrations and is a direct noblotting, distinguishes between natural infection and vacci-
result of FIV infection (rather than the result of opportunistic nation or the presence of maternal antibody. Kittens that test
infection). Total protein concentrations that ranged from 4.5 to positive should be retested after 6 months of age (the Advisory
11 g/dL were reported in one study.68 Other findings are vari- Bureau on Cat Diseases recommends retesting after 4 months
able and relate to the presence of concurrent disease, neoplasia, of age).66,73 Nevertheless, kittens less than 6 months of age
or opportunistic infections. should still be tested, because the vast majority of negative
kittens will be declared free of infection. Molecular testing
Urinalysis could be considered to confirm infection in kittens that test
Urinalysis may reveal proteinuria in cats with glomerulonephritis. positive at less than 6 months of age (see later discussion).
Cats with a history of FIV vaccination may remain antibody-
Coagulation Testing positive for more than 4 years.66 Because infection can occur
Cats infected with FIV can have mild prolongations in their in the face of vaccination, positive test results in a vaccinated
APTT, thrombin time, and fibrinogen concentrations, although cat may represent infection and/or historical vaccination.
the PT and platelet function are generally normal.69 Currently, molecular testing with PCR assays is required to
identify infection in these cats, but some infected cats test
Cytologic Evaluation of Bone Marrow PCR-negative. An ELISA assay has been developed that can
Bone marrow cytologic evaluation in FIV-infected cats with distinguish naturally infected from vaccinated cats, but this
cytopenias and nonregenerative anemias may show mild dys- assay is not commercially available. The assay was used to
plasia (usually not as severe as in FeLV infections); erythroid test blood samples from 73 uninfected, unvaccinated cats; 89
hypoplasia; and/or myeloid hyperplasia despite peripheral leu- uninfected, FIV-vaccinated cats; and 102 FIV-infected cats,
kopenia, sometimes with a left shift. The latter suggests ineffec- including 3 cats that had been vaccinated.9 The assay had a
tive hematopoiesis or maturation arrest. sensitivity of 97% and a specificity of 100% for detection of
FIV infection in these cats.
Microbiologic Tests False-negative ELISA assay results occur early in the course
Serologic Diagnosis of illness, because cats may take up to 60 days to develop an
The initial assay of choice for diagnosis of, and screening for, antibody response.66 Rarely, antibody production is delayed
FIV infection is an ELISA assay that detects antibody to FIV. for 6 months or longer. Thus when recent exposure is possible,
Provided there has not been a history of vaccination for FIV testing should be repeated a minimum of 2 months later. False-
and the tested cat is less than 6 months of age, positive anti- negative test results also occur in cats in the terminal phase
body test results equate with infection, because the virus estab- of disease, as a result of impaired antibody production, or in
lishes a lifelong, persistent infection. Point-of-care, lateral-flow kittens with rapidly progressive infections. These cats often
ELISA assays and diagnostic laboratory-based ELISA assays have high plasma viral loads. Thus, if advanced FIV infection
are in widespread use and have rapid turnaround times and is suspected, negative test results should be followed by virus
high sensitivities and specificities.70 These assays usually detect detection using PCR. When serology is used as a screening test,
antibodies to the FIV p24 core protein, and sometimes to the negative results are considered to be highly reliable because of
gp40 transmembrane protein. False positive test results occur the high sensitivity of the test and the low prevalence of infec-
rarely as a result of operator error or nonspecific reactivity tion in most populations of healthy cats.66
against tissue culture components after vaccination.71 When Immunofluorescent antibody (IFA) assays that detect anti-
Western immunoblotting was used as the gold standard, the bodies to FIV have also been described. When performed by
sensitivities, specificities, and positive and negative predic- experience laboratory personnel, these have sensitivities and
tive values of six different ELISA assays (Synbiotics Witness, specificities that range from 95% to 100% when compared with
Viracheck FIV, IDEXX SNAP Combo Plus, PetChek Plus Western immunoblotting.71
Anti-FIV, MegaCor Fastest, and Bio Veto Duo Speed) in one
U.S. study of 535 cats with an overall FIV seroprevalence of Molecular Diagnosis Using the Polymerase Chain Reaction
10.3% ranged from 94.5% to 100%, 98.5% to 100%, 91.2% A variety of PCR assays have been developed for diagnosis of
to 100%, and 99.2% to 100%, respectively.70 The Mapic FIV FIV infection. Assays may detect viral RNA (reverse transcrip-
test had a high (23.1%) rate of invalid test results and was tase [RT]-PCR), proviral DNA, or both RNA and proviral DNA
not recommended for clinical practice. When these assays are in peripheral blood. Because the FIV vaccine is inactivated and
216 SECTION 1  Viral Diseases

A B
FIGURE 21-5  A, Depletion of paracortical areas in the mesenteric lymph node of an 8-year-old female spayed domestic shorthair with FIV infection that was euthanized as a result of
the development of progressive neurologic signs. B, Normal feline lymph node for comparison purposes.

does not replicate or integrate into the host genome, PCR assays expensive and can take 2 to 3 weeks, so it is not used for routine
should not detect vaccine virus in cats that have been vacci- diagnostic purposes.
nated for FIV. Compared with serology, PCR can be insensitive
(sensitivity <80%), because viral loads in healthy cats are often Pathologic Findings
extremely low, and some strains may not be detected because Gross pathologic findings in cats with FIV infection include
of variability in the sequence of the viral genome among FIV emaciation, stomatitis, lymphadenopathy, and evidence of
isolates. Sensitivity is likely to be higher in cats in the acute secondary neoplasia or opportunistic infections. Evidence of
and terminal phases of disease when viral loads are higher, but disorders of aged cats, such as hyperthyroidism, cardiomy-
this requires further study. False-positive test results have the opathy, and interstitial nephritis, may also be present. Histo-
potential to occur as a result of laboratory contamination; in pathologic changes in cats with FIV infection include follicular
some commercial laboratories, unacceptable sensitivities and hyperplasia or, in the chronic phase, lymphoid depletion in
specificities have been reported (as low as 41% and 44%, the paracortical regions and plasma cell infiltration of lymph
respectively).74,75 In advertising materials, IDEXX Laborato- nodes (Figure 21-5); lymphoplasmacytic ulcerative stomatitis
ries report an assay sensitivity and specificity of 80.5% and (see Figure 21-4, B); and bone marrow pathology as described
99.9%, respectively, among 36 FIV-infected cats, 96 uninfected previously in this chapter. Evidence of chronic interstitial
vaccinated cats and 92 uninfected unvaccinated cats, with no nephritis, sometimes with membranous glomerulonephritis
difference in specificity in the latter two groups.76 A FRET real- and glomerulosclerosis, may be present. Inflammatory changes
time PCR assay directed at the gag gene has been described may be seen within skeletal muscle and/or the gastrointesti-
that differentiates between FIV subtypes (see Chapter 5 for an nal tract. Histopathology of the CNS in cats infected with
explanation of FRET PCR assays).11 The assay was positive in neurovirulent FIV strains may show mild lymphoplasmacytic
60% of 101 cats with positive antibody tests for FIV. A history meningitis, perivascular lymphocytic infiltrates, diffuse gliosis,
of vaccination with the FIV vaccine was present in 13 of the microglial nodules, and mild neuronal degeneration and apop-
cats, and all these cats tested negative. The vaccination history tosis.59,78 Neuronal dysfunction may be more important than
of the remaining cats was unknown. Given their limitations, neuronal loss.59
PCR assays should not be used for diagnosis of FIV infection
in the absence of concurrent serologic testing, and the results Treatment and Prognosis
of PCR assays should not be used to decide whether vaccina-
tion should be performed. Cats that are seronegative that test Supportive Care
positive with PCR may be in the terminal phase of disease and Cats in the terminal phase of FIV infection may require fluid
unable to produce antibody, or the PCR assay result may be a therapy, nutritional support, regular dental prophylaxis, dilute
false positive. Latently infected cats have rarely been described chlorhexidine-based mouth washes or oral gels, and dental
that test positive for proviral DNA but negative for antibody.76 extractions and antimicrobial drugs with activity against anaer-
These cats lack any immunologic or clinical abnormalities that obes for severe stomatitis (Table 21-5). Whole mouth extrac-
occur with active infection.76 Whether these cats are likely to tions have been beneficial for some cats with severe stomatitis,
develop productive infection (as reported in humans with latent but it is critical that all tooth roots be removed. Referral to a
HIV infection) is unknown.77 board-certified veterinary dentist should be considered. Topi-
cal glucocorticoids (e.g., 1% prednisolone acetate, every 4 to
Virus Isolation 12 hours depending on severity) and topical atropine are indi-
FIV can be isolated from peripheral blood lymphocytes in pri- cated for cats with anterior uveitis. Systemic glucocorticoids
mary feline T cell cultures, but this is technically demanding and should only be used if absolutely necessary, because their use
CHAPTER 21  Feline Immunodeficiency Virus Infection 217

TABLE 21-5
Suggested Medications for Treatment of Cats in the Terminal Phase of Feline Immunodeficiency Virus Infection
Drug Dose Route Interval (hours) Comments
Zidovudine (AZT) 5 to 15 mg/kg PO 12 May have benefit for cats with neurologic signs
or stomatitis. Monitor CBC (see text). Toxicity
may be more likely at doses above 5 mg/kg.
Human recombinant 1 to 50 U/cat PO 24 For stomatitis
interferon alpha
0.12% Chlorhexidine 1 mL Topical 12 to 48 For stomatitis
plus zinc gluconate
oral gel
Clavulanic acid­– 12.5 to 25 mg/kg PO 12 For stomatitis
amoxicillin
Lactoferrin 200 mg powder or Topical 24 For refractory gingivitis and stomatitis. Must be
40 mg/kg solution purchased from chemical suppliers.
Prednisolone 5 mg PO 24 For refractory stomatitis or advanced neurologic
disease. May also be required for lymphoma or
other CNS neoplasms. Use only if absolutely
necessary.

is associated with increased plasma viremia. Some cats with study, a decreased proviral load was reported without evidence
advanced neurologic signs show clinical improvement after of adverse effects, but there was no improvement in clinical or
treatment with glucocorticoids. Opportunistic infections may immunologic variables.73,83
respond to appropriate antimicrobial treatment, but prolonged Results of treatment with human recombinant IFN-α have
or lifelong treatment may be required. Griseofulvin should not been mixed, but prolonged survival was reported in 24 FIV-
be used to treat dermatophytosis, because it has been associated infected cats after oral administration of a low dose of the drug
with bone marrow suppression in FIV-positive cats.79 Recom- when compared with 6 untreated control cats.83 Clinical scores
binant human erythropoietin or darbepoetin may be useful for and laboratory parameters improved in some sick, naturally
some cats with nonregenerative anemia and does not appear infected cats that were treated with recombinant feline IFN-ω,
to increase viral load through activation of transcription of despite no significant changes in viral load.84
latent virus.80 Although recombinant human granulocyte col-
ony stimulating factor (G-CSF) can increase neutrophil counts Management of FIV-Infected Cats
in cats infected with FIV, antibodies can develop within a few Cats that test positive for FIV should be housed indoors to
weeks that cross-react with endogenous G-CSF. When recombi- prevent transmission to other cats, as well as to protect them
nant human granulocyte-macrophage colony stimulating factor from other infections. The latter is important even in subclini-
(GM-CSF) was used, an increase in virus load occurred, so the cally infected cats, as other infections have the potential to acti-
use of these drugs is not recommended. vate viral transcription and accelerate disease progression. The
feeding of raw foods and hunting behavior should be avoided.
Antiviral and Immunomodulator Drugs Cats should be neutered to reduce the chance of roaming and
Topical lactoferrin administration has been associated with aggressive interactions with other cats. Minimally, FIV-posi-
clinical improvement in some FIV-infected cats with stomatitis tive cats should be rechecked every 6 months in order to moni-
(see Chapter 7). Cats with stomatitis or neurologic disease may tor body weight, assess for periodontal disease, and discuss the
benefit from oral or subcutaneous treatment with zidovudine need for routine laboratory testing and vaccination with core
(AZT), but AZT can cause bone marrow suppression, so the vaccines. A complete physical examination, CBC, serum bio-
CBC must be monitored weekly for the first month and monthly chemistry panel, and urinalysis are recommended on an annual
thereafter. Treatment is not recommended for cats with myelo- basis.66 At least during acute FIV infection, cats can mount
suppressive disease.73 AZT should be discontinued if the hema- an immune response to vaccines,85,86 but because vaccination
tocrit drops below 20%, after which anemia usually resolves in with core vaccines may activate viral transcription, vaccination
a few days.73 AZT-resistant FIV mutants can develop as early as should be performed only for FIV-positive cats that are likely
6 months after the start of treatment. Fozivudine (45 mg/kg PO to be exposed to other cats. The use of inactivated vaccines
q12h) reduces viremia in cats with acute FIV infection and may is recommended, because of the potential for vaccine-induced
be less likely to produce hematologic adverse effects.81 Plerixa- disease in immunosuppressed cats.73 When hospitalized, FIV-
for (AMD3100), a selective antagonist of the CXCR4 recep- infected cats should be kept in separate cages away from
tor, is active against FIV in vitro.82 When used in 40 naturally isolation, where there may be other cats with transmissible
infected cats for 6 weeks in a placebo-controlled double-blind infectious diseases. Perioperative antimicrobial drug treatment
218 SECTION 1  Viral Diseases

could be considered for cats that undergo invasive surgery or were not protected from challenge with FIV subtype A strain
dental treatments.66,73 Glasgow-8; all vaccinated and control cats were infected.92
Because of diagnostic test interference, uncertain efficacy in
Prognosis the field, and the increased risk of sarcoma formation associ-
A limited number of studies have shown no significant difference ated with adjuvanted vaccines, the FIV vaccine is considered a
in life span between FIV-infected cats and uninfected cats.42,87 noncore vaccine that should only be used for cats at high risk
In one study, the median survival times of these two groups of infection, such as outdoor cats that fight with other cats. The
after testing for FIV infection were 3.9 (39 cats) and 5.9 (22 initial vaccine should only be given to cats that test negative
cats) years, respectively.87 In a larger study of more than 1000 with FIV ELISA assays. Cat owners should be informed that
FIV-infected cats and more than 8000 age- and sex-matched protection is incomplete and that identification of subsequent
control cats, the median survival times of the two groups were infection may be impossible in some cats with the available diag-
4.9 years and 6.0 years, respectively.66 The progression and nostic tests. The initial vaccine dose is followed by two booster
severity of disease is related to virus strain and host immunity. doses at 3- to 4-week intervals, followed by annual boosters
Infection of geriatric and neonatal cats is associated with more so long as risk persists. An identification microchip should be
rapid progression and severity of disease than infection of young placed that links FIV vaccination to the microchip number, so
adult cats.84,88 For example, progression to terminal immuno- should the cat escape or be relinquished to a shelter, a positive
deficiency can occur within 2 months in kittens.89 Because the antibody test result is not interpreted as FIV infection.
life span of FIV-infected cats may not differ greatly from that
of uninfected cats, no cat should be euthanized on the basis of Prevention
a positive FIV test alone.66,73 However, other factors, such as
control of the infection in group-housed or breeding cats, may Transmission of FIV is reduced when cats are housed indoors.
necessitate euthanasia or rehoming of some cats that test posi- When one positive cat is identified in a household, all other
tive. Once terminal FIV-related disease occurs, life spans are cats should be tested and no new cats should be introduced,
typically less than 1 year. as this may lead to conflict and increased fighting behavior.
Isolation of positive cats in a household should be considered.
Immunity and Vaccination In breeding catteries, infection can be prevented by screening
all new introductions, preferably with a 2-month quarantine
The FIV vaccine first became available in the United States in period for introductions followed by retesting, and removal
July 2002 (Fel-O-Vax FIV, Fort Dodge Animal Health) and is of all infected queens and toms from the cattery. Transmis-
an inactivated, adjuvanted, whole-virus vaccine that contains sion in shelters can be reduced by testing on intake or housing
FIV Petaluma (subtype A) and FIV Shizuoka (subtype D). The cats singly in cages, and testing at or shortly after adoption,
vaccine has been available in New Zealand and Australia since together with education of adopters in regard to the need to
2004. It is licensed for the vaccination of healthy cats that are 8 retest for infection.
weeks of age or older as an aid in the prevention of FIV, and in Surgical needles, endotracheal tubes, breathing circuits, and
experimental challenge studies, protected 60% to 80% of cats instruments should never be shared between cats without proper
from infection. The introduction of the vaccine generated con- cleaning and disinfection, and all cats used as blood donors
troversy, because (1) existing serologic assays cannot differenti- should test negative for FIV antibodies with ELISA and, if pos-
ate between natural infection and vaccination, (2) vaccination sible, PCR assays offered by a diagnostic laboratory that has
provides only partial protection from infection, and (3) PCR a high level of quality control. Fluid lines and multidose vials
assay results cannot be relied upon in vaccinated cats. Although should not be shared between cats, because they can become
concerns exist that the vaccine may only protect against infec- contaminated with body fluids.
tion with strains from homologous subtypes, studies have shown
moderate protection against infection with subtype B strains.90,91 Public Health Aspects
In one study, 10 of 14 cats were protected from subtype B infec-
tion when challenged 1 year after vaccination compared with Despite its similarities to HIV, no evidence of natural infection
none of 5 control cats.90 However, 13-week-old vaccinated cats of humans with FIV exists.
CHAPTER 21  Feline Immunodeficiency Virus Infection 219

CASE EXAMPLE Laboratory Findings:


CBC:
HCT 22.5% (30%-50%)
Signalment: “Arthur” an 11-year-old male neutered domestic MCV 47.5 fL (42-53 fL)
shorthair from northern California MCHC 32.9 g/dL (30-33.5 g/dL)
History: Arthur was initially brought to his veterinarian for a Reticulocytes 7400 cells/µL
2-day history of lethargy and decreased appetite. Physical WBC 1100 cells/µL (4500-14,000 cells/µL)
examination was unremarkable. Blood work showed a Neutrophils 704 cells/µL (2000-9000 cells/µL)
hematocrit of 28% (reference range, 29% to 38%), a white Band neutrophils 99 cells/µL
cell count of 6400 cells/µL with a normal differential, Lymphocytes 220 cells/µL (1000-7000 cells/µL)
clumped platelets, mild hyperbilirubinemia (0.5 mg/dL), Monocytes 77 cells/µL (50-600 cells/µL)
mild hypoalbuminemia (2.4 g/dL), and hyperglobulinemia 32,000 platelets/µL (180,000-500,000 platelets/µL).
(5.5 g/dL). He was treated with amoxicillin and mirtazapine, Neutrophils showed moderate toxicity with many Döhle
but lethargy and inappetence persisted. Serial hemograms bodies, and there were many macroplatelets.
performed over the subsequent 3 months showed Serum Chemistry Profile:
progressive leukopenia due to a neutropenia and Sodium 149 mmol/L (151-158 mmol/L)
lymphopenia (Table 21-6), during which time Arthur was Potassium 3.1 mmol/L (3.6-4.9 mmol/L)
treated with orbifloxacin, with some improvement in his Chloride 115 mmol/L (117-126 mmol/L)
appetite and attitude. A bone marrow aspirate performed 5 Bicarbonate 17 mmol/L (15-21 mmol/L)
weeks after the onset of illness showed myeloid hyperplasia Phosphorus 4.3 mg/dL (3.2-6.3 mg/dL)
with a left shift. Serology for FIV and FeLV was performed Calcium 8.8 mg/dL (9.0-10.9 mg/dl)
11 weeks after onset of illness, and the cat was negative BUN 17 mg/dL (18-33 mg/dL)
for FeLV antigen and had an equivocal test result for FIV Creatinine 1.1 mg/dL (1.1-2.2 mg/dL)
antibody. Serum thyroxine concentration was low (0.7 mcg/ Glucose 158 mg/dL (63-118 mg/dL)
dL, reference range 1.1 to 3.3 mcg/dL). The cat was referred Total protein 8.1 g/dL (6.6-8.4 g/dL)
for further evaluation. Albumin 3.0 g/dL (2.2-4.6 g/dL)
Arthur was an indoor and outdoor cat and had a history of pre- Globulin 5.1 g/dL (2.8-5.4 g/dL)
dation (mice and birds) and fighting with other cats in the ALT 121 U/L (27-101 U/L)
neighborhood. His diet otherwise consisted of a commercial AST 113 U/L (17-58 U/L)
wet cat food. He was vaccinated regularly for rabies, FeLV, ALP 10 U/L (14-71 U/L)
feline herpesvirus-1, feline calicivirus, and feline panleuko- Creatine kinase 278 U/L (73-260 U/L)
penia virus infections. Gamma GT <3 U/L (0-4 U/L)
Current Medications: Orbifloxacin, 7 mg/kg PO q12h Cholesterol 204 mg/dL (89-258 mg/dL)
Other Medical History: Routine blood work had been Total bilirubin 0.8 mg/dL (0-0.2 mg/dL)
performed 3 years previously, which showed a normal CBC, Magnesium 2.4 mg/dL (1.5-2.5 mg/dL).
a globulin of 5 g/dL, a urine specific gravity of 1.069, and a T4 Anti-nuclear Antibody Serology: Positive at 1:32
of 1.3. At that time, Arthur tested negative for FeLV antigen Microbiologic and Virologic Testing: Point-of-care ELISA
and positive for FIV antibody. serology for FeLV antigen and FIV antibody: negative
Physical Examination: Feline coronavirus antibody serology: negative at 1:25
Body Weight: 3.1 kg Aerobic and anaerobic blood cultures (three specimens): negative
General: Quiet, alert, responsive, estimated to be 5% to 7% PCR for FIV proviral DNA: positive (cycle threshold value = 30,
dehydrated, T = 103.9° F (39.9° C), HR = 144 beats/min, values <40 are positive)
RR = 50 breaths/min, mucous membranes pale pink, CRT Imaging Findings:
< 2 s. A dry haircoat with moderate amounts of scale was Abdominal Ultrasound: Hepatomegaly was identified and
present. There was no evidence of ectoparasites. the spleen had a mottled echotexture. There was mild
Eyes, Ears, Nose, and Throat (with Dilated Fundoscopic enlargement of the ileocecal lymph nodes and mild diffuse
Examination): Moderate gingivitis and absent mandibular thickening of the small intestinal submucosal layer.
canine teeth. No other clinically significant abnormalities Thoracic Radiographs: Cardiopulmonary structures were
were detected. within normal limits.
Musculoskeletal: Body condition score 2/9. Generalized Cytologic Findings:
muscle atrophy was noted. Ambulatory in all four limbs. Liver Aspiration Cytology: A moderately bloody background
Cardiovascular: Aside from an intermittent gallop rhythm, no contained scattered, variably sized clusters of uniform
abnormalities were noted. hepatocytes and low numbers of nucleated cells. The nucleated
Respiratory: No abnormalities were detected. cells consisted of a mixture of nondegenerate neutrophils,
Gastrointestinal and Urogenital: Abdomen soft and lymphocytes, macrophages, and rare myeloid precursors.
nonpainful on palpation, mild hepatomegaly. Urinary Several macrophages were erythrophagic. Interpretation:
bladder was small and soft. Possible mild myeloid extramedullary hematopoiesis.
Lymph Nodes: All lymph nodes less than 1 cm in diameter. Spleen Aspiration Cytology: The specimen was highly cellular
with a pink background that contained a moderate amount
220 SECTION 1  Viral Diseases

of blood and scattered clumps of hemosiderin. A mixed marked erythroid hypoplasia, and moderate megakaryocytic
population of lymphocytes was admixed with variable hypoplasia. The myeloid hyperplasia together with persistent
numbers of hematopoietic precursor cells. Hematopoietic neutropenia suggested myelodysplasia.
precursors were primarily granulocytic precursors, with Serum Protein Electrophoresis: A mild increase in α2
many progranulocytes. A few erythroid precursors, including globulins was present (1.34 g/dL, reference range, 0.4-0.9
erythroblasts, were noted. Plasma cells were moderately g/dL) together with a broad-based gamma region. This was
increased in number, and both immature and mature forms consistent with an acute-phase inflammatory response.
were seen. Several binucleated and rare trinucleated plasma Diagnosis: Terminal phase of FIV infection, characterized by
cells and scattered immature lymphocytes were noted. pancytopenia secondary to myelodysplasia; hepatopathy
Vacuolated macrophages were increased in number and (open diagnosis).
often contained phagocytized erythrocytes or blue-green Comments: The progression of FIV infection was apparent
pigment consistent with hemosiderin. Scattered mitotic in this cat, which transitioned from a seropositive to a
figures were noted. Interpretation: Moderate plasmacytosis, seronegative, PCR-positive state as a result of progressive
extramedullary hematopoiesis, and histiocytic hyperplasia decline in immune function and, ultimately, failure to
with erythrophagocytosis and increased iron. The large produce antibodies. The cause of the hepatopathy was
number of immature plasma cells raised the possibility of not identified. Blood cultures were negative, and spleen
plasma cell neoplasia; however, reactive plasmacytosis could and liver aspiration cytology failed to reveal evidence of
not be ruled out. underlying neoplasia. Because of the plasmacytosis in the
Bone Marrow Aspiration Cytology: Numerous hypercellular spleen, serum protein electrophoresis was performed to
unit particles were present. A few mature megakaryocytes assist diagnosis of a plasma cell tumor (as supported by a
were noted. Hematopoietic cells consisted largely of myeloid monoclonal gammopathy), but this was not apparent. The
precursors, with an estimated myeloid to erythroid ratio cat was treated with clavulanic acid–amoxicillin without
greater than 10. Myeloid cells showed orderly maturation to clinical improvement. Treatment with glucocorticoids was
the band neutrophil stage, with few segmented neutrophils. also offered because of the possibility of undiagnosed
Few erythroid precursors were noted, which included rare round cell neoplasia or immune-mediated neutropenia (in
prorubricytes, rubricytes, and metarubricytes. Scattered well- light of the myeloid hyperplasia combined with a positive
differentiated plasma cells and a small amount of hemosiderin antinuclear antibody test). The cat died at home 1 month
were noted. Interpretation: Marked myeloid hyperplasia, later, and necropsy was not performed.

TABLE 21-6
Progression of Laboratory Abnormalities in an 11-Year-old Male Neutered Domestic Shorthair That Tested Positive
for Antibodies to FIV 3 Years Earlier, at Which Time a CBC was Normal

Days after Onset of Illness


39 47 56 74 83 Reference Range
Hematocrit (%) 29 29 33 27 22 30-50
White blood cells/µL 2000 2100 1000 1100 1100 4500-14,000
Neutrophils/µL 1180 1407 610 572 704 2000-9000
Lymphocytes/µL 700 588 340 462 220 1000-7000
Platelets/µL 191,000 196,000 Clumped Clumped 32,000 180,000-500,000
FIV/FeLV serology* Equivocal/ Negative/
negative negative
*SNAP FIV/FeLV Combo Test, IDEXX Laboratories.

SUGGESTED READINGS Levy J, Crawford C, Hartmann K, et al. 2008 American Association of


Feline Practitioners’ feline retrovirus management guidelines. J Feline
Goldkamp CE, Levy JK, Edinboro CH, et  al. Seroprevalences of feline Med Surg. 2008;10:300-316.
leukemia virus and feline immunodeficiency virus in cats with abscesses
or bite wounds and rate of veterinarian compliance with current guide- REFERENCES
lines for retrovirus testing. J Am Vet Med Assoc. 2008;232:1152-1158.
Hartmann K. Clinical aspects of feline immunodeficiency and feline leuke- 1. Pedersen NC, Ho EW, Brown ML, et al. Isolation of a T-lympho-
mia virus infection. Vet Immunol Immunopathol. 2011;143(3-4):190- tropic virus from domestic cats with an immunodeficiency-like syn-
201 (for a review of molecular pathogenetic mechanisms of disease). drome. Science. 1987;235:790-793.
Hosie MJ, Addie D, Belak S, et  al. Feline immunodeficiency. ABCD 2. Gruffydd-Jones TJ, Hopper CD, Harbour DA, et  al. Serological
guidelines on prevention and management. J Feline Med Surg. evidence of feline immunodeficiency virus infection in UK cats from
2009;11:575-584. 1975-76. Vet Rec. 1988;123:569-570.
CHAPTER 21  Feline Immunodeficiency Virus Infection 221

3. Reid RW, Barr MC, Scott FW. Retrospective serologic survey for 24. Martins AN, Medeiros SO, Simonetti JP, et  al. Phylogenetic and
the presence of feline immunodeficiency virus antibody: a compari- genetic analysis of feline immunodeficiency virus gag, pol, and env
son of ELISA and IFA techniques. Cornell Vet. 1992;82:359-369. genes from domestic cats undergoing nucleoside reverse transcrip-
4. Troyer JL, Pecon-Slattery J, Roelke ME, et  al. Seroprevalence tase inhibitor treatment or treatment-naive cats in Rio de Janeiro,
and genomic divergence of circulating strains of feline immuno- Brazil. J Virol. 2008;82:7863-7874.
deficiency virus among Felidae and Hyaenidae species. J Virol. 25. Kelly PJ, Stocking R, Gao D, et al. Identification of feline immuno-
2005;79:8282-8294. deficiency virus subtype-B on St. Kitts, West Indies by quantitative
5. Elder JH, Lin YC, Fink E, et  al. Feline immunodeficiency virus PCR. J Infect Dev Ctries. 2011;5:480-483.
(FIV) as a model for study of lentivirus infections: parallels with 26. Hayward JJ, Taylor J, Rodrigo AG. Phylogenetic analysis of feline
HIV. Curr HIV Res. 2010;8:73-80. immunodeficiency virus in feral and companion domestic cats of
6. de Parseval A, Chatterji U, Sun P, et al. Feline immunodeficiency New Zealand. J Virol. 2007;81:2999-3004.
virus targets activated CD4+ T cells by using CD134 as a binding 27. Nakamura K, Suzuki Y, Ikeo K, et al. Phylogenetic analysis of Viet-
receptor. Proc Natl Acad Sci U S A. 2004;101:13044-13049. namese isolates of feline immunodeficiency virus: genetic diversity
7. Shimojima M, Miyazawa T, Ikeda Y, et  al. Use of CD134 as a of subtype C. Arch Virol. 2003;148:783-791.
primary receptor by the feline immunodeficiency virus. Science. 28. Uema M, Ikeda Y, Miyazawa T, et  al. Feline immunodeficiency
2004;303:1192-1195. virus subtype C is prevalent in northern part of Taiwan. J Vet Med
8. Samman A, McMonagle EL, Logan N, et al. Phylogenetic charac- Sci. 1999;61:197-199.
terisation of naturally occurring feline immunodeficiency virus in 29. Reggeti F, Bienzle D. Feline immunodeficiency virus subtypes A, B
the United Kingdom. Vet Microbiol. 2011;150:239-247. and C and intersubtype recombinants in Ontario, Canada. J Gen
9. Hayward JJ, Rodrigo AG. Molecular epidemiology of feline immu- Virol. 2004;85:1843-1852.
nodeficiency virus in the domestic cat (Felis catus). Vet Immunol 30. Pecoraro MR, Tomonaga K, Miyazawa T, et al. Genetic diversity
Immunopathol. 2010;134:68-74. of Argentine isolates of feline immunodeficiency virus. J Gen Virol.
10. Weaver EA. A detailed phylogenetic analysis of FIV in the United 1996;77(Pt 9):2031-2035.
States. PLoS One. 2010;5:e12004. 31. Nichols J, Litster A, Leutenegger C. Relationship between viral
11. Wang C, Johnson CM, Ahluwalia SK, et al. Dual-emission fluores- subgroup, viral load and health status in a population of cats natu-
cence resonance energy transfer (FRET) real-time PCR differentiates rally infected with feline immunodeficiency virus. Proceedings of
feline immunodeficiency virus subtypes and discriminates infected the 2nd Symposium of the International Society for Companion
from vaccinated cats. J Clin Microbiol. 2010;48:1667-1672. Animal Infectious Diseases. 2012:Abstract 07.
12. Nakamura Y, Ura A, Hirata M, et al. An updated nation-wide epi- 32. Ueland K, Nesse LL. No evidence of vertical transmission of natu-
demiological survey of feline immunodeficiency virus (FIV) infec- rally acquired feline immunodeficiency virus infection. Vet Immu-
tion in Japan. J Vet Med Sci. 2010;72:1051-1056. nol Immunopathol. 1992;33:301-308.
13. Bachmann MH, Mathiason-Dubard C, Learn GH, et  al. Genetic 33. Allison RW, Hoover EA. Covert vertical transmission of feline immu-
diversity of feline immunodeficiency virus: dual infection, recom- nodeficiency virus. AIDS Res Hum Retroviruses. 2003;19:421-434.
bination, and distinct evolutionary rates among envelope sequence 34. Jordan HL, Howard J, Barr MC, et  al. Feline immunodeficiency
clades. J Virol. 1997;71:4241-4253. virus is shed in semen from experimentally and naturally infected
14. Steinrigl A, Klein D. Phylogenetic analysis of feline immunodefi- cats. AIDS Res Hum Retroviruses. 1998;14:1087-1092.
ciency virus in Central Europe: a prerequisite for vaccination and 35. Jordan HL, Howard JG, Bucci JG, et al. Horizontal transmission
molecular diagnostics. J Gen Virol. 2003;84:1301-1307. of feline immunodeficiency virus with semen from seropositive cats.
15. Kann R, Seddon J, Kyaw-Tanner M, et al. Phylogenetic analysis to J Reprod Immunol. 1998;41:341-357.
define feline immunodeficiency virus subtypes in 31 domestic cats 36. Little S, Sears W, Lachtara J, et al. Seroprevalence of feline leuke-
in South Africa. J S Afr Vet Assoc. 2006;77:108-113. mia virus and feline immunodeficiency virus infection among cats
16. Kann RK, Kyaw-Tanner MT, Seddon JM, et al. Molecular subtyp- in Canada. Can Vet J. 2009;50:644-648.
ing of feline immunodeficiency virus from domestic cats in Austra- 37. Gleich SE, Krieger S, Hartmann K. Prevalence of feline immuno-
lia. Aust Vet J. 2006;84:112-116. deficiency virus and feline leukaemia virus among client-owned
17. Kann RK, Seddon JM, Meers J, et  al. Feline immunodeficiency cats and risk factors for infection in Germany. J Feline Med Surg.
virus subtypes in domestic cats in New Zealand. N Z Vet J. 2009;11:985-992.
2007;55:358-360. 38. Goldkamp CE, Levy JK, Edinboro CH, et  al. Seroprevalences of
18. Kann R, Seddon J, Kyaw-Tanner M, et al. Co-infection with dif- feline leukemia virus and feline immunodeficiency virus in cats
ferent subtypes of feline immunodeficiency virus can compli- with abscesses or bite wounds and rate of veterinarian compliance
cate subtype assignment by phylogenetic analysis. Arch Virol. with current guidelines for retrovirus testing. J Am Vet Med Assoc.
2007;152:1187-1193. 2008;232:1152-1158.
19. Iwata D, Holloway SA. Molecular subtyping of feline immunodefi- 39. Levy JK, Edinboro CH, Glotfelty CS, et  al. Seroprevalence of
ciency virus from cats in Melbourne. Aust Vet J. 2008;86:385-389. Dirofilaria immitis, feline leukemia virus, and feline immunode-
20. Sodora DL, Shpaer EG, Kitchell BE, et  al. Identification of three ficiency virus infection among dogs and cats exported from the
feline immunodeficiency virus (FIV) env gene subtypes and com- 2005 Gulf Coast hurricane disaster area. J Am Vet Med Assoc.
parison of the FIV and human immunodeficiency virus type 1 evo- 2007;231:218-225.
lutionary patterns. J Virol. 1994;68:2230-2238. 40. Levy JK, Scott HM, Lachtara JL, et  al. Seroprevalence of feline
21. Kakinuma S, Motokawa K, Hohdatsu T, et al. Nucleotide sequence leukemia virus and feline immunodeficiency virus infection among
of feline immunodeficiency virus: classification of Japanese isolates cats in North America and risk factors for seropositivity. J Am Vet
into two subtypes which are distinct from non-Japanese subtypes. Med Assoc. 2006;228:371-376.
J Virol. 1995;69:3639-3646. 41. Yamamoto JK, Hansen H, Ho EW, et al. Epidemiologic and clini-
22. Pistello M, Cammarota G, Nicoletti E, et al. Analysis of the genetic cal aspects of feline immunodeficiency virus infection in cats from
diversity and phylogenetic relationship of Italian isolates of feline the continental United States and Canada and possible mode of
immunodeficiency virus indicates a high prevalence and heteroge- transmission. J Am Vet Med Assoc. 1989;194:213-220.
neity of subtype B. J Gen Virol. 1997;78(Pt 9):2247-2257. 42. Addie DD, Dennis JM, Toth S, et al. Long-term impact on a closed
23. Nishimura Y, Goto Y, Pang H, et al. Genetic heterogeneity of env household of pet cats of natural infection with feline coronavirus,
gene of feline immunodeficiency virus obtained from multiple dis- feline leukaemia virus and feline immunodeficiency virus. Vet Rec.
tricts in Japan. Virus Res. 1998;57:101-112. 2000;146:419-424.
222 SECTION 1  Viral Diseases

43. Hellard E, Fouchet D, Santin-Janin H, et  al. When cats’ ways of 63. O’Neil LL, Burkhard MJ, Hoover EA. Frequent perinatal transmis-
life interact with their viruses: a study in 15 natural populations sion of feline immunodeficiency virus by chronically infected cats.
of owned and unowned cats (Felis silvestris catus). Prev Vet Med. J Virol. 1996;70:2894-2901.
2011;101:250-264. 64. Weaver CC, Burgess SC, Nelson PD, et  al. Placental immunopa-
44. Yamamoto JK, Sparger E, Ho EW, et  al. Pathogenesis of experi- thology and pregnancy failure in the FIV-infected cat. Placenta.
mentally induced feline immunodeficiency virus infection in cats. 2005;26:138-147.
Am J Vet Res. 1988;49:1246-1258. 65. English RV, Davidson MG, Nasisse MP, et al. Intraocular disease
45. Tompkins MB, Tompkins WA. Lentivirus-induced immune dys- associated with feline immunodeficiency virus infection in cats.
regulation. Vet Immunol Immunopathol. 2008;123:45-55. J Am Vet Med Assoc. 1990;196:1116-1119.
46. Mexas AM, Fogle JE, Tompkins WA, et al. CD4+CD25+ regula- 66. Levy J, Crawford C, Hartmann K, et al. 2008 American Associa-
tory T cells are infected and activated during acute FIV infection. tion of Feline Practitioners’ feline retrovirus management guide-
Vet Immunol Immunopathol. 2008;126:263-272. lines. J Feline Med Surg. 2008;10:300-316.
47. Vahlenkamp TW, Tompkins MB, Tompkins WA. Feline immuno- 67. Goto Y, Nishimura Y, Baba K, et al. Association of plasma viral
deficiency virus infection phenotypically and functionally activates RNA load with prognosis in cats naturally infected with feline
immunosuppressive CD4+CD25+ T regulatory cells. J Immunol. immunodeficiency virus. J Virol. 2002;76:10079-10083.
2004;172:4752-4761. 68. Gleich S, Hartmann K. Hematology and serum biochemistry of
48. Dean GA, LaVoy A, Yearley J, et al. Cytokine modulation of the feline immunodeficiency virus-infected and feline leukemia virus-
innate immune response in feline immunodeficiency virus-infected infected cats. J Vet Intern Med. 2009;23:552-558.
cats. J Infect Dis. 2006;193:1520-1527. 69. Hart SW, Nolte I. Hemostatic disorders in feline immunodeficiency
49. Lehman TL, O’Halloran KP, Hoover EA, et  al. Utilizing the FIV virus-seropositive cats. J Vet Intern Med. 1994;8:355-362.
model to understand dendritic cell dysfunction and the potential 70. Hartmann K, Griessmayr P, Schulz B, et  al. Quality of different
role of dendritic cell immunization in HIV infection. Vet Immunol in-clinic test systems for feline immunodeficiency virus and feline
Immunopathol. 2010;134:75-81. leukaemia virus infection. J Feline Med Surg. 2007;9:439-445.
50. Levy JK, Liang Y, Ritchey JW, et  al. Failure of FIV-infected cats 71. Barr MC, Pough MB, Jacobson RH, et al. Comparison and inter-
to control Toxoplasma gondii correlates with reduced IL2, IL6, pretation of diagnostic tests for feline immunodeficiency virus
and IL12 and elevated IL10 expression by lymph node T cells. Vet infection. J Am Vet Med Assoc. 1991;199:1377-1381.
Immunol Immunopathol. 2004;98:101-111. 72. Levy JK, Crawford PC, Slater MR. Effect of vaccination against
51. Fletcher NF, Meeker RB, Hudson LC, et al. The neuropathogenesis feline immunodeficiency virus on results of serologic testing in cats.
of feline immunodeficiency virus infection: barriers to overcome. Vet J. J Am Vet Med Assoc. 2004;225:1558-1561.
2011;188:260-269. 73. Hosie MJ, Addie D, Belak S, et al. Feline immunodeficiency. ABCD
52. Hofmann-Lehmann R, Berger M, Sigrist B, et al. Feline immunode- guidelines on prevention and management. J Feline Med Surg.
ficiency virus (FIV) infection leads to increased incidence of feline 2009;11:575-584.
odontoclastic resorptive lesions (FORL). Vet Immunol Immuno- 74. Bienzle D, Reggeti F, Wen X, et al. The variability of serological
pathol. 1998;65:299-308. and molecular diagnosis of feline immunodeficiency virus infection.
53. Beatty JA, Troyer RM, Brewester C, et  al. Feline immunodefi- Can Vet J. 2004;45:753-757.
ciency virus (FIV)-associated lymphoma. Is a gammaherpesvirus 75. Crawford PC, Slater MR, Levy JK. Accuracy of polymerase chain
involved?. Proceedings of the 2nd Symposium of the International reaction assays for diagnosis of feline immunodeficiency virus
Society for Companion Animal Infectious Diseases. 2012:Abstract infection in cats. J Am Vet Med Assoc. 2005;226:1503-1507.
03. 76. Dandekar S, Beebe AM, Barlough J, et al. Detection of feline immu-
54. Magden E, Quackenbush SL, VandeWoude S. FIV associ- nodeficiency virus (FIV) nucleic acids in FIV-seronegative cats.
ated neoplasms—a mini-review. Vet Immunol Immunopathol. J Virol. 1992;66:4040-4049.
2011;143:227-234. 77. IDEXX Reference Laboratories Diagnostic Update. 2009. http://
55. Beatty JA, Callanan JJ, Terry A, et  al. Molecular and immuno- www.idexx.com/view/xhtml/en_us/smallanimal/reference-
phenotypical characterization of a feline immunodeficiency virus laboratories/testmenu/innovative-tests/real-pcr.jsf?SSOTOKEN=0.
(FIV)-associated lymphoma: a direct role for FIV in B-lymphocyte Last accessed May 18, 2012.
transformation?. J Virol. 1998;72:767-771. 78. Maingat F, Vivithanaporn P, Zhu Y, et  al. Neurobehavioral per-
56. Matsumoto H, Takemura N, Sako T, et al. Serum concentration of formance in feline immunodeficiency virus infection: integrated
circulating immune complexes in cats infected with feline immuno- analysis of viral burden, neuroinflammation, and neuronal injury
deficiency virus detected by immune adherence hemagglutination in cortex. J Neurosci. 2009;29:8429-8437.
method. J Vet Med Sci. 1997;59:395-396. 79. Shelton GH, Grant CK, Linenberger ML, et al. Severe neutropenia
57. Fujino Y, Horiuchi H, Mizukoshi F, et al. Prevalence of hemato- associated with griseofulvin therapy in cats with feline immunode-
logical abnormalities and detection of infected bone marrow cells ficiency virus infection. J Vet Intern Med. 1990;4:317-319.
in asymptomatic cats with feline immunodeficiency virus infection. 80. Arai M, Darman J, Lewis A, et al. The use of human hematopoi-
Vet Microbiol. 2009;136:217-225. etic growth factors (rhGM-CSF and rhEPO) as a supportive ther-
58. Shelton GH, Linenberger ML, Grant CK, et  al. Hematologic apy for FIV-infected cats. Vet Immunol Immunopathol. 2000;77:
manifestations of feline immunodeficiency virus infection. Blood. 71-92.
1990;76:1104-1109. 81. Fogle JE, Tompkins WA, Campbell B, et al. Fozivudine tidoxil as
59. Meeker RB. Feline immunodeficiency virus neuropathogenesis: single-agent therapy decreases plasma and cell-associated viremia
from cats to calcium. J Neuroimmune Pharmacol. 2007;2:154-170. during acute feline immunodeficiency virus infection. J Vet Intern
60. Podell M, Chen E, Shelton GD. Feline immunodeficiency Med. 2011;25:413-418.
virus associated myopathy in the adult cat. Muscle Nerve. 82. Egberink HF, De Clercq E, Van Vliet AL, et al. Bicyclams, selec-
1998;21:1680-1685. tive antagonists of the human chemokine receptor CXCR4,
61. Maingat F, Halloran B, Acharjee S, et al. Inflammation and epithe- potently inhibit feline immunodeficiency virus replication. J Virol.
lial cell injury in AIDS enteropathy: involvement of endoplasmic 1999;73:6346-6352.
reticulum stress. FASEB J. 2011;25:2211-2220. 83. Hartmann K, Stengel C, Klein D, et al. Efficacy and adverse effects
62. O’Neil LL, Burkhard MJ, Diehl LJ, et al. Vertical transmission of of the antiviral compound pleraxifor in feline immunodeficiency
feline immunodeficiency virus. Semin Vet Med Surg (Small Anim). virus-infected cats. J Vet Intern Med. 2012;26(3):483-490.
1995;10:266-278.
CHAPTER 21  Feline Immunodeficiency Virus Infection 223

84. Pedersen NC, Leutenegger CM, Woo J, et al. Virulence differences 89. O’Neil LL, Burkhard MJ, Obert LA, et  al. Regression of feline
between two field isolates of feline immunodeficiency virus (FIV- immunodeficiency virus infection. AIDS Res Hum Retroviruses.
APetaluma and FIV-CPGammar) in young adult specific pathogen 1997;13:713-718.
free cats. Vet Immunol Immunopathol. 2001;79:53-67. 90. Huang C, Conlee D, Gill M, et  al. Dual-subtype feline immu-
85. Doménech A, Miró G, Collado VM, et  al. Use of recombinant nodeficiency virus vaccine provides 12 months of protective
interferon omega in feline retrovirosis: from theory to practice. Vet immunity against heterologous challenge. J Feline Med Surg.
Immunol Immunopathol. 2011;143(3-4):301-306. 2010;12:451-457.
86. Dawson S, Smyth NR, Bennett M, et  al. Effect of primary-stage 91. Kusuhara H, Hohdatsu T, Okumura M, et al. Dual-subtype vac-
feline immunodeficiency virus infection on subsequent feline calici- cine (Fel-O-Vax FIV) protects cats against contact challenge
virus vaccination and challenge in cats. AIDS. 1991;5:747-750. with heterologous subtype B FIV infected cats. Vet Microbiol.
87. Ravi M, Wobeser GA, Taylor SM, et al. Naturally acquired feline 2005;108:155-165.
immunodeficiency virus (FIV) infection in cats from western Can- 92. Dunham SP, Bruce J, MacKay S, et  al. Limited efficacy of an
ada: Prevalence, disease associations, and survival analysis. Can inactivated feline immunodeficiency virus vaccine. Vet Rec.
Vet J. 2010;51:271-276. 2006;158:561-562.
88. George JW, Pedersen NC, Higgins J. The effect of age on the course
of experimental feline immunodeficiency virus infection in cats.
AIDS Res Hum Retroviruses. 1993;9:897-905.
CHAPTER 22

Feline Leukemia Virus Infection


Jane E. Sykes and Katrin Hartmann

FeLV-C. Each subtype uses a different receptor to enter cells


Overview of Feline Leukemia Virus Infection (Table 22-1). All cats infected with FeLV-B and FeLV-C are co-
First Described: Scotland, 1964 (Jarrett et al.)1 infected with FeLV-A, and only FeLV-A is transmitted between
Cause: Feline leukemia virus (family Retroviridae, subfamily animals. FeLV-B and FeLV-C are more pathogenic than FeLV-
Orthoretrovirinae, genus Gammaretrovirus) A. FeLV-B arises through recombination of FeLV-A proviral
DNA with endogenous FeLV sequences present in host cellu-
Affected Hosts: Domestic and some wild Felidae
lar DNA.3 FeLV-C arises from accumulation of mutations or
Geographic Distribution: Worldwide insertions in the env (SU) gene of FeLV-A.4,5 The FeLV subtype
Mode of Transmission: Prolonged close contact with salivary influences the clinical expression of disease (see Table 22-1).
secretions; to a lesser extent biting, transplacental trans- For example, FeLV-C is associated with nonregenerative ane-
mission, transmission through milk, and through blood mia. Even within an FeLV subtype, mutations in the SU and
transfusion the LTR regions of the viral genome affect disease outcome.6,7
An additional subtype, FeLV-T, has been associated with
Major Clinical Signs: Lethargy, fever, pallor, stomatitis, signs immunodeficiency.
of underlying lymphoma or leukemia, signs of immune- Transmission of FeLV-A primarily results from close con-
mediated disorders or opportunistic infections tact with salivary secretions, such as through licking, mutual
Differential Diagnoses: FIV infection is the primary differen- grooming, and shared food and water dishes. Other routes of
tial diagnosis for cats with signs of immunosuppression; transmission, such as by biting, blood transfusion, in milk, and
feline calicivirus infection, or FIV infection are differen- possibly by fleas, can also occur.8-10 The virus survives poorly
tial diagnoses for cats with stomatitis; primary immune- outside the cat and is readily inactivated by disinfectants, soap,
mediated disease and other bone marrow diseases are and desiccation. The overall prevalence of FeLV infection
differential diagnoses for cats with immune-mediated has declined over the past two decades with more extensive
diseases and cytopenias testing and vaccination. Before the institution of widespread
Human Health Significance: FeLV does not infect humans. testing and vaccination, more than 30% of cats in some catter-
ies were infected.11 In the early 1990s, the overall prevalence
of infection was 13% in nearly 28,000 sick cats from North

Etiology and Epidemiology


Matrix
FeLV is an enveloped RNA virus that belongs to the genus Gam-
Capsid protein (p27)
maretrovirus of the family Retroviridae. FeLV infection remains
an important cause of mortality in domestic cats through its
ability to cause immune suppression, bone marrow disorders,
and hematopoietic neoplasia. FeLV also causes disease in wild
felids such as the highly endangered Iberian lynx.2 Integrase
FeLV infection progresses more rapidly than FIV infection
and is more pathogenic, so most cats that develop progressive Reverse transcriptase
infections ultimately die of FeLV-related disease. However, in
contrast to FIV infection, many cats in the early state of FeLV
infection regress to a permanent state of viral latency (“regres-
sive infection”). It is possible that some cats, after exposure RNA
to a low dose of FeLV, may eliminate the infection altogether
(“abortive infection”), although this appears to be a rare out-
come. Regardless, a positive test result for FeLV infection in an Surface glycoprotein (SU or gp70)
apparently healthy cat does not always imply that FeLV-related FIGURE 22-1  Structure of feline leukemia virus. Gammaretroviruses contain two
disease and mortality will occur. identical strands of RNA and associated enzymes, which include reverse transcriptase,
The structure of FeLV is similar to that of FIV, except that integrase, and protease, packaged into a core composed of the capsid protein (p27) with a
the capsid is icosahedral rather than cone shaped (Figure 22-1). surrounding matrix, all enclosed by a phospholipid membrane envelope derived from the
There are three main subtypes of FeLV: FeLV-A, FeLV-B, and host cell. The envelope contains a gp70 glycoprotein and the transmembrane protein p15E.

224
CHAPTER 22  Feline Leukemia Virus Infection 225

TABLE 22-1
Host Cellular Receptors Involved in FeLV Infection
FeLV Subtype Receptor Receptor Function Comments
FeLV-A FeTHTR1* Thiamine transporter protein Present in all cats with FeLV; transmitted exog-
enously
FeLV-B FePit1 or FePit2† Inorganic phosphate transporter Results from recombination between FeLV-A
protein and feline endogenous FeLV-related retrovirus
sequences; may accelerate development of lym-
phoma or enhance ­neuropathogenicity
FeLV-C FLVCR‡ Heme transporter protein Arises from point mutations in FeLV-A env gene;
­associated with nonregenerative anemia

*Mendoza R, Anderson MM, Overbaugh J. A putative thiamine transport protein is a receptor for feline leukemia virus subgroup A. J Virol
2006;80(7):3378-3385.
†Anderson MM, Lauring AS, Robertson S, et al. Feline Pit2 functions as a receptor for subgroup B feline leukemia viruses. J Virol 2001;75(22):

10563-10572.
‡Keel SB, Doty RT, Yang Z, et al. A heme export protein is required for red blood cell differentiation and iron homeostasis. Science 2008;319(5864):

825-828.

America that were at risk for exposure.12 The prevalence was as fever, lethargy, and/or lymphadenopathy, during this period.
just 7% in a similar population of approximately 1400 cats A small number of infected lymphocytes then travel to the bone
in 2006.13 The prevalence of FeLV infection in European cats marrow, where the virus infects rapidly dividing precursor cells
has also declined markedly.14 Currently, the overall prevalence and subsequently lymphoid and epithelial cells throughout the
of infection in mixed populations of cats is 1% to 6%.13,15-19 body. This infection of the bone marrow is considered a critical
Cats with access to the outdoors; those that have contact with step in the pathogenesis of FeLV infection. Once infection of
other cats; cats that are male, aggressive, or intact; and cats epithelial cells in the salivary glands occurs, the virus is shed in
that are co-infected with FIV are at increased risk of FeLV massive quantities in saliva; low quantities of virus can also be
infection.12,13,15,17 Male cats are less strongly predisposed to shed in urine and feces.
FeLV infection than to FIV infection, and in some studies,20 Possible outcomes of infection with FeLV are shown in
no male predisposition has been recognized. Adult cats are Figure 22-2. The immune system of some infected cats sup-
more likely to be infected with FeLV than cats aged less than 6 presses viral replication within a few weeks after infection,
months,13,17 but the median age of cats infected with FeLV is before significant infection of the marrow occurs. These cats
3 years,15 which is lower than that for FIV. This reflects a) the develop a regressive infection, whereby proviral DNA is present
greater degree of pathogenicity of FeLV than FIV and its ability in the host cell genome but production and shedding of virus no
to significantly reduce lifespan; and b) the phenomenon of age- longer occurs (see Chapter 21). This may occur after the initial
related resistance to FeLV, whereby exposure of cats that are period of viremia, or viremia may never be detectable.22 Regres-
less than 4 months of age to the virus is much more likely to lead sive infection can persist for life and may be reactivated with
to progressive infection than exposure of adults.21 When 12- to immunosuppression, such as might occur during pregnancy or
16-week old kittens are exposed to a multicat household with following treatment with immunosuppressive drugs.23 Later in
endemic FeLV infection, between 60% and 70% of kittens will life, an unknown percentage of cats with regressive infections
become infected within a 5-month time period. In contrast, less may develop FeLV-negative malignancies as a result of integra-
than 5% of cats aged 6 months or older will become infected tion of viral DNA within host cellular oncogenes. Most cats
over the same time period; over a 2-year period, 40% to 50% with regressive infection, however, never develop clinical signs
of these adult cats will become infected. Infection of adult cats related to FeLV infection. Viral genome sequences may eventu-
can also still occur with exposure to high doses of the virus or ally be incompletely replicated, and as a consequence, reactiva-
when there is underlying host immune compromise. tion of virus replication will become impossible in some cats
over time. In abortive infections, no viremia occurs after infec-
Clinical Features tion, and virus cannot be detected using any method. Cats with
abortive infections have been exposed to low doses of FeLV,
Signs and Their Pathogenesis and although they fail to develop viremia, they will develop
The outcome of FeLV infection is extremely variable and antibodies to the virus.24 Some cats that never test antigen-
depends strongly on the virus strain involved, the challenge positive may have focal infections, that is, evidence of proviral
dose, the route of inoculation, and factors that influence host DNA in some tissues but not in blood or bone marrow.9 Cats
immune function such as age, genetics, co-infections, stress, develop progressive infection once involvement of the marrow
and treatment with immunosuppressive drugs. After oronasal is established and cellular destruction by the virus exceeds the
exposure to the virus, the virus replicates in oral lymphoid tis- ability of the host’s immune system to suppress viral replica-
sue and then circulates in a few monocytes and lymphocytes tion. Persistent viremia and progressive FeLV-related disease
within peripheral blood. Some cats develop systemic signs, such result (Box 22-1).
226 SECTION 1  Viral Diseases

FeLV exposure
All antigen tests –
BOX 22-1
All PCR tests –
Clinical Outcomes of Progressive FeLV Infection
Week 1
Variable
Neoplasia, especially lymphoma, leukemia and fibrosarco-
Strong
immune response immune response mas (with FeSV)
Opportunistic infections
Abortive infection Focal/Atypical infection Pure red cell aplasia
Virus eliminated Antigen – or transiently + Aplastic anemia, myelodysplasia, or myelofibrosis
All antigen tests – Blood PCR – Immune-mediated disease (immune-mediated hemolytic
All PCR tests – Tissue proviral PCR + anemia, thrombocytopenia, glomerulonephritis, polyar-
Week 3 thritis, uveitis)
Peripheral lymphadenopathy
p27 ELISA antigen +
IFA antigen – Neurologic disease (e.g., anisocoria, urinary incontinence)
Good All PCR tests + Poor/Absent Reproductive failure
immune response immune response Gastrointestinal disease (uncommon)
Other (e.g., osteochondromatosis, cutaneous horns)
Regressive infection Progressive infection
All antigen tests – All antigen tests + FeSV, feline sarcoma virus.
Proviral PCR Weak + All PCR tests +
RT-PCR – Immunosuppression

FeLV-related disease
FIGURE 22-2  Outcomes of infection with FeLV. In the first week of infection, cats
test negative with all antigen assays (i.e., p27 ELISA and IFA assays), RT-PCR for viral RNA,
and PCR for proviral DNA. Rarely, cats exposed to low levels of virus eliminate the infec-
tion before or at this time point (abortive infection). Soluble antigen assays and proviral
PCR assays become positive from about the third week after infection, and virus is shed
in saliva, tears, urine, and feces. After this time point, productive viral infection is sup-
pressed by the host immune response (regressive infection) or the virus multiplies rapidly
in the bone marrow and a progressive infection occurs, which is characterized by persis-
tently positive antigen ELISA, IFA, viral RNA, and proviral DNA assay results. Cats with focal A
or atypical infection are never or transiently antigen positive and test negative using PCR
assays on blood or bone marrow, but some tissues (such as the mammary gland or blad-
der) are positive with proviral PCR. The latter occurs in only a small percentage (e.g., <
5%) of infected cats.

Opportunistic Infections
Opportunistic infections may develop in FeLV infections as a
result of myelo suppression or an acquired cell-mediated immu-
nodeficiency. The immunosuppressive properties of FeLV are
not fully understood but have been linked in part to the viral
envelope peptide, p15E, which inhibits T and B cell function,
inhibits cytotoxic lymphocyte responses, alters monocyte mor-
phology and distribution, and has been associated with impaired
cytokine production and responsiveness.25-28 Kittens with pro-
gressive FeLV infection have impaired T cell and, to a lesser B
extent, B cell function.29-32 Infected cats may develop lymphope-
nia, thymic atrophy, and depletion of lymphocytes within lymph FIGURE 22-3  Opportunistic infections in cats with progressive FeLV infection. A,
node paracortical zones. CD4+ T cell malfunction may contrib- Ulceration of the nasal planum in a 6-month-old male neutered domestic medium hair
ute to a decreased humoral and cell-mediated immune response cat with progressive FeLV infection as defined by positive ELISA antigen and IFA assays
on peripheral blood. Stomatitis was also present, and feline calicivirus infection was sus-
in affected cats,33,34 and the response to vaccination may also be
pected. B, Severe nasal cryptococcosis in a Siamese cat from a cattery with endemic FeLV
impaired. Impaired neutrophil function compounds the effect of infection.
neutropenia in some infected cats.35-37 Opportunistic infections
that result include bacterial infections of the upper and lower
urinary tract, hemoplasmosis, upper respiratory tract infections, but clinical signs are more severe and refractory to therapy.
feline infectious peritonitis, chronic stomatitis, toxoplasmosis, Infection with FeLV is an established risk factor for hemoplasma
dermatophytosis, and cryptococcosis (Figure 22-3). The preva- infection. In one study, it was also a risk factor for Bartonella
lence of some of these infections in cats with FeLV infection does henselae infection, but no evidence of clinical disease was identi-
not differ significantly from that in cats not infected with FeLV, fied in the cats infected with B. henselae.38
CHAPTER 22  Feline Leukemia Virus Infection 227

Neoplasia
FeLV causes neoplasia in cats primarily as a result of insertional BOX 22-2
mutagenesis, by which the virus activates proto-oncogenes (espe-
cially c-myc, but also others such as flit-1) or disrupts tumor
Mechanisms of Anemia in Cats Infected with FeLV
suppressor genes.39 The most common types of neoplasia in cats
infected with FeLV are lymphoma and leukemia. Cats infected Decreased RBC Production
with FeLV are more than 60-fold more likely to develop lym- Pure red cell aplasia (FeLV-C)
phoma than cats not infected with FeLV; this compares with Aplastic anemia
approximately 5-fold for FIV.40 Lymphoma or leukemia can Leukemia (myelophthisis)
be detected in nearly one quarter of cats with progressive FeLV Myelofibrosis
infection at necropsy.41 In the 1980s, as many as 70% of cases of Anemia of inflammatory disease
feline lymphoma were associated with FeLV infection, but with
improved control measures and vaccination, the vast majority RBC Loss
of cats (more than 80% to 90%) seen at veterinary clinics with Thrombocytopenia secondary to immune-mediated
lymphoma now test negative for FeLV antigen.42-44 The most or bone marrow disease
common types of lymphoma in cats infected with FeLV are
thymic (mediastinal), multicentric, spinal, renal, or ocular lym- Increased RBC Destruction
phoma. FeLV-associated lymphomas are mostly of T-cell origin, FeLV-associated IMHA
but B cell lymphoma can also occur. In contrast, FIV-associated Co-infection with hemoplasmas
lymphomas tend to be of B-cell origin.42 Approximately 80%
of cats with thymic lymphoma test positive for FeLV antigen, IMHA, immune-mediated hemolytic anemia.
whereas fewer than 10% of cats with gastrointestinal lymphoma
are FeLV antigen-positive. Large granular lymphoma is rarely
associated with FeLV infection.45 Cats with thymic lymphoma
typically develop clinical signs of lethargy, tachypnea, and some- sites with a poor prognosis. FeLV-FeSV DNA sequences have
times regurgitation. Most FeLV-positive cats with lymphoma are been detected in some uveal melanomas from cats,55 but other
less than 4 years of age.14 studies have failed to identify an association between FeSV and
Cats with regressive infection appear to be at greater risk for ocular melanomas or sarcomas in cats.56,57 FeLV infection does
development of lymphoma than cats that were never exposed not appear to be important in the pathogenesis of solitary fibro-
to FeLV. FeLV-negative cats that live with cats that test posi- sarcomas or injection-site sarcomas in cats.58,59
tive for FeLV antigen have a more than 40-fold increased risk Other tumor types that are variably associated with FeLV
of lymphoma compared to that expected without exposure infection are feline olfactory neuroblastomas, cutaneous horns,
to FeLV.46 Several studies have investigated the prevalence of and osteochondromatosis (multiple cartilaginous exostoses).60
FeLV proviral DNA in tumor cells from cats that test negative Feline olfactory neuroblastomas are rare and aggressive tumors
for FeLV antigen. Some studies (including more recent studies of the nasal cavity that can lead to signs of rhinosinusitis and
that used real-time PCR assays) found no evidence of FeLV pro- neurologic signs. Osteochondromatosis is characterized by
viral DNA in feline lymphomas.14,47,48 In older studies that used benign cartilage-capped exostotic growths that arise from the
conventional PCR, more than 20% of antigen-negative lym- surface of a bone, which can cause pain, lameness, disfigure-
phomas tested positive.49,50 Additional studies from a variety ment, and paresis due to spinal cord compression. Cutaneous
of geographic locations that use multiple real-time PCR assays horns occur as a result of benign keratinocyte hyperplasia.
are required to clarify the role that FeLV plays in lymphomas
and leukemias that develop in cats that test negative for FeLV Anemia and Bone Marrow Disorders
antigen. Multiple mechanisms can lead to anemia in cats infected with
FeLV is responsible for the majority of myelogenous leuke- FeLV (Box 22-2). Approximately 90% of FeLV-associated ane-
mias, erythroleukemias, megakaryocytic, and lymphoid leuke- mias are nonregenerative.61 A variety of bone marrow disorders
mias in cats, although not all cats with these tumors test positive lead to decreased red blood cell production. FeLV-C infection
for FeLV antigen. Most FeLV-associated leukemias are acute. results in pure red cell aplasia, a severe nonregenerative ane-
FeLV infection can underlie chronic eosinophilic leukemia,51 mia associated with erythrocyte macrocytosis and depletion of
chronic myelomonocytic leukemia,52 and chronic lymphocytic erythroid precursors in the bone marrow. This occurs because
leukemia. However, most cats with chronic lymphocytic leuke- FeLV-C binds to and interferes with a heme exporter protein,
mia are seronegative for FeLV antigen. which results in subsequent heme toxicosis to the developing
FeLV infection can result in the development of multiple erythrocyte.62 The presence of macrocytosis in the absence of
fibrosarcomas in young cats. These occur when FeLV-A recom- reticulocytosis should thus raise suspicion for FeLV infection.
bines with cellular oncogenes (such as c-fes, c-fms, or c-fgr) to FeLV infection can also lead to aplastic anemia in some cats,
form feline sarcoma viruses (FeSV). These viruses then develop which is a deficiency in all cell lineages (platelets, myeloid, and
mutations in these oncogenes that, when reinserted into cellular erythroid) in the bone marrow, and replacement of the bone
DNA, cause malignant transformation.53,54 To replicate, FeSV marrow space by adipose tissue. Anemia and bone marrow
requires the presence of FeLV-A, which supplies proteins such as dysfunction may also result from myelophthisis secondary to
those encoded by the env gene. Thus, all cats infected with FeSV leukemia, myeloid and/or erythroid dysplasia, or myelofibrosis
test positive for FeLV antigen. FeSV-associated fibrosarcomas (progressive replacement of the marrow with collagenous con-
are characterized by multifocal, locally invasive, often ulcerated nective tissue) (Figure 22-4). Myelodysplasia is characterized by
cutaneous masses that metastasize readily to the lung and other disordered maturation of marrow precursors, which in some
228 SECTION 1  Viral Diseases

most common form of MDS in cats is MDS-EB.64 Regressive


FeLV infections have been detected by proviral DNA PCR in
some cats with bone marrow disorders,65 but the relationship
between the presence of proviral DNA and the pathogenesis of
disordered marrow function is unclear.
Anemia in cats with FeLV infection may also result from
anemia of inflammatory disease, which can be triggered by
opportunistic infections or neoplastic disease. Erythrocyte
destruction occurs in some FeLV-infected cats as a result of
secondary immune-mediated hemolytic anemia (IMHA) or co-
infection with hemoplasmas (see Chapter 41). Hemorrhage as
a result of thrombocytopenia or defective platelet function may
also contribute to anemia. Mechanisms of thrombocytopenia
include immune-mediated thrombocytopenia (ITP) and bone
marrow dysfunction. Similarly, neutropenia can result from
myeloid hypoplasia, myelofibrosis, myelodysplasia, myelo-
A phthisis, or maturation arrest at the myelocyte or metamyelo-
cyte stages. In some cases, peripheral neutropenia accompanied
by myeloid hyperplasia reflects underlying immune-mediated
neutropenia.

Immune-Mediated Disorders
In addition to immune-mediated cytopenias, other immune-
mediated disorders that can occur in association with progressive
FeLV infection are glomerulonephritis,66 uveitis,67 and polyar-
thritis.68 Circulating immune complexes have been detected in
infected cats.69-71 Because primary immune-mediated disorders
are otherwise rare in cats, retrovirus testing is essential in cats
that are diagnosed with these conditions before immunosuppres-
sive drug treatment is initiated.

Neurologic Disorders
Neurologic disorders in cats with progressive FeLV infections
B may occur secondary to central nervous system (CNS) neo-
FIGURE 22-4  Bone marrow abnormalities in cats with FeLV infection. A, Bone mar- plasia, opportunistic infections, or FeLV infection itself. The
row aspirate cytology from a 6-month old male neutered domestic shorthair cat that was envelope protein of FeLV may be neurotoxic.72,73 Anisocoria,
evaluated for lethargy and fever. Acute leukemia, marked erythroid aplasia, marked gran- mydriasis, Horner’s syndrome, and urinary incontinence can
ulocytic dysplasia, and increased eosinophilopoiesis is present. Megakaryocytic dysplasia occur. FeLV infection may be one of the most frequent causes
was also identified. The G:E ratio was approximately 2950:1 with marked erythroid aplasia. of urinary incontinence in cats, an otherwise uncommon condi-
Wright’s stain. B, Bone marrow aspirate cytology from a 1-year-old male neutered domes- tion. A myelopathy has been described in FeLV-infected cats
tic shorthair. Severe megaloblastic erythrodysplasia and mild megakaryocytic dysplasia that showed various neurologic signs such as disorientation,
were identified. The myeloid series is present but markedly decreased in all stages; the
lethargy, increased vocalization, progressive ataxia, paresis,
myeloid to erythroid ratio was estimated at 1:6.5 (compare with A). Nuclear to cytoplasmic
asynchrony (megaloblastic change) is present, with immature nuclei in cytoplasm with paralysis, hyperesthesia, urinary incontinence, recurrent consti-
hemoglobinization. Mitotic figures are present in moderate numbers and are seen in the pation, and anisocoria with diminished pupillary light reflexes.74
late stages of the erythroid series (arrows). At necropsy, abundant FeLV antigen was detectable within neu-
rons, endothelial cells, oligodendroglia, and astrocytes within
the spinal cord.74
cases precedes emergence of leukemia. FeLV infection under-
lies many myelodysplastic syndromes (MDS) and leukemia in Gastrointestinal Disease
cats.63 However, over the past two decades, 50% of 28 cats Uncommonly, FeLV causes enteritis that clinically and histo-
with myelodysplasia and 44% of 41 cats with leukemia seen logically resembles that caused by feline panleukopenia virus
at the University of California, Davis, tested negative for FeLV (FPV), except that lymphoid depletion is absent.75 Clinical signs
antigen in peripheral blood, and only 36% of 34 cats with dys- include vomiting, acute or chronic diarrhea that may be hemor-
myelopoiesis seen at the University of Minnesota were positive rhagic, inappetence, weight loss, and dehydration. FeLV-infected
for FeLV antigen.64 Several different classification systems have cats with “panleukopenia-like syndrome” have intestinal crypt
been used to describe feline MDS.64 The French-American- destruction and pancytopenia that results from myeloid destruc-
British (FAB) classification scheme used for humans has been tion (Figure 22-5).76 Although now very rare, co-infections with
modified for dogs and cats. This divides MDS into 2 groups: FeLV and FPV can also occur, and the 2 viruses may enhance
1) MDS with refractory cytopenia (less than 6% myeloblasts each other’s pathogenicity.77 Co-infections with FeLV and other
in the bone marrow, MDS-RC) and 2) MDS with excessive enteric viruses, such as feline coronavirus, also occur.78 In most
numbers of myeloblasts (6% to 30% myeloblasts, MDS-EB). cases, diarrhea in cats infected by FeLV results from an etiology
The presence of more than 30% blasts indicates leukemia. The other than FeLV alone.
CHAPTER 22  Feline Leukemia Virus Infection 229

Diagnosis
Infection with FeLV is often diagnosed when healthy cats are
screened for infection. Screening should be performed with
ELISA or related immunochromatographic in-house assays for
free FeLV antigen in serum, because these assays are sensitive,
specific, rapid, widely available, and most well understood. The
retrovirus status of all cats should be known regardless of the
presence of absence of illness.79 The indications for retrovirus
testing are described in Chapter 21 (see Box 21-1).
Even though many cats that test positive for FeLV antigen
have no clinical signs or physical examination abnormalities, a
CBC, chemistry panel, and urinalysis should be obtained from
these cats (and at a minimum, a complete CBC with blood smear
evaluation) to assess for underlying abnormalities that could sig-
nal the presence of FeLV-related disorders. Subtle hematologic
FIGURE 22-5  Histopathology of a colonic biopsy specimen from a 6-year-old intact abnormalities, such as erythroid macrocytosis or monocytope-
male FeLV+ domestic shorthair with a 1-week history of anorexia, weight loss, lethargy, nia, may be present in the absence of overt clinical signs and can
fever, and bloody mucoid diarrhea. Severe, diffuse, plasmacytic colitis was present with signify a poorer long-term outcome. Additional diagnostic tests
crypt epithelial necrosis and regeneration. One crypt is dilated, lined with attenuated indicated in infected cats that are anemic include a reticulocyte
epithelium, and contains necrotic debris (red arrow). An adjacent crypt shows evidence count, Coombs’ test, and PCR assay for hemoplasmas. Bone
of regeneration (blue arrow). A tissue gram stain showed a mixed population of bacteria marrow aspiration and core biopsy are indicated in cats with
on the luminal surface, but not within crypts. A CBC showed thrombocytopenia (22,000 pancytopenia or persistent nonregenerative anemias.
platelets/µL) and neutropenia (2700 neutrophils/µL) with neutrophil toxicity. A fecal par-
vovirus antigen test was negative, and bone marrow aspirate cytology showed evidence of Laboratory Abnormalities
myelodysplasia. Remarkably, this cat lived another 3 years with transfusion support, but
ultimately was euthanized because of acute granulocytic leukemia with secondary sepsis. Complete Blood Count
The CBC may be normal or show regenerative or nonregenera-
tive anemia, neutropenia, lymphopenia, monocytopenia, and/
or thrombocytopenia. Evidence of agglutination may be pres-
ent in cats with IMHA. Moderate to marked leukocytosis and
Reproductive Disorders increased band neutrophils may also be present. Large numbers
Transplacental spread of FeLV can lead to fetal resorption, abor- of circulating blasts, megakaryocytes or dysplastic cells (such
tion, neonatal death, and fading kitten syndrome.76 Fetal loss as erythrocytes with giant Howell-Jolly bodies) can be found in
may also result from secondary endometritis. Kittens infected in cats with leukemia or MDS. When compared with uninfected
late gestation or after birth develop thymic atrophy, failure to cats, FeLV-infected cats were nearly 3.8-fold more likely to be
nurse, dehydration, lethargy, and death within the first 2 weeks anemic, 5-fold more likely to be thrombocytopenic, 3.6-fold
of life. more likely to be neutropenic, and 2.8-fold more likely to have
lymphocytosis.80
Physical Examination Findings
Physical examination findings in cats with progressive FeLV Serum Biochemical Tests and Urinalysis
infection vary dramatically depending on the stage of infec- Findings on serum biochemistry analysis and urinalysis are
tion and the secondary disease process present. There may be nonspecific and reflect underlying disease processes. Hyper-
no abnormalities, or cats may show signs of lethargy, pyrexia, bilirubinemia and bilirubinuria may be present in cats with
mucosal pallor, petechial hemorrhages, dehydration, peripheral immune-mediated hemolytic anemia or hemoplasmosis. Cats
lymphadenomegaly, thin body condition, stomatitis, subcutane- with glomerulonephritis may be proteinuric. Some cats have
ous abscesses, or upper respiratory tract disease. Anemic cats evidence of bacterial urinary tract infections. Urine culture and
may have a hemic murmur, tachypnea, or tachycardia, or they susceptibility testing of a urine specimen obtained via cystocen-
may be icteric. Cats with severe anemia can be laterally recum- tesis are indicated in cats with suspected urinary tract infection.
bent or comatose and hypothermic. Cats with thymic lymphoma
may be tachypneic and have decreased lung and cardiac sounds Bone Marrow Cytology and Histopathology
on thoracic auscultation as a result of malignant pleural effusion, Both bone marrow aspirate and core biopsy specimens should
or heart sounds may be displaced caudally. Decreased compress- be obtained in cats with pancytopenia or nonregenerative ane-
ibility of the cranial thorax may also be detected. Splenomegaly, mia. If aspirate results are not diagnostic, the core biopsy should
hepatomegaly, renomegaly, intestinal masses, and/or abdominal be submitted for interpretation. This is because bone marrow
lymph node enlargement may be detected on abdominal palpa- aspirates from cats with aplastic anemia or myelofibrosis are
tion of cats with multicentric lymphoma. Neurologic signs are typically of low cellularity. Bone marrow findings in cats with
detected infrequently when compared with signs of anemia or FeLV infection include evidence of neoplastic lymphoid, ery-
thoracic or abdominal lymphoma and include ataxia, anisoco- throid, or myeloid cells (which circulate in the peripheral blood
ria, and mydriasis. Although also relatively uncommon, uveitis of cats with leukemia); myelodysplasia; hypoplasia or aplasia
may be identified, or there may be other ocular abnormalities of erythroid, myeloid, or megakaryocyte cell lines; erythroid,
as a result of intraocular lymphoma or co-infections with other myeloid, and megakaryocyte hyperplasia despite peripheral
pathogens. cytopenias; and megakaryocyte hypoplasia. Cytochemical stains
230 SECTION 1  Viral Diseases

that identify cells of the myeloid lineage (such as alkaline phos- on thoracic radiography that may be accompanied by mild to
phatase, peroxidase, Sudan black B, and nonspecific esterase), severe pleural effusion (Figure 22-6). Abdominal sonography in
immunocytochemistry, or flow cytometry using antibodies that cats with multicentric lymphoma may reveal hypoechoic and
target cell surface cluster of differentiation (CD) molecules may enlarged abdominal lymph nodes and enlargement, hypoecho-
be needed to definitively identify the cell type involved in some genicity, or mottling of the spleen, liver, or kidneys. Increased
acute undifferentiated leukemias. hepatic echogenicity can also occur with lymphoma. Intestinal
masses with loss of normal bowel wall layering may also be
Diagnostic Imaging detected. Splenomegaly may be detected in cats with immune-
Imaging findings in cats with FeLV infection reflect the underly- mediated cytopenias.
ing disease process and are extremely variable. Cats with FeLV-
associated thymic lymphosarcoma have a mediastinal mass Microbiologic Tests
Diagnostic assays available for FeLV infection are listed in
Table 22-2.

Antigen Assays
The initial assay of choice for diagnosis of FeLV infection is an
ELISA or a similar immunochromatographic test that detects
soluble p27 capsid protein antigen in blood. The term soluble is
used to distinguish these assays from assays such as IFA, which
detect fixed antigen within cells. In most cats, the presence of
circulating antigen correlates with viremia, although a few cats
have viremia in the absence of detectable antigen or antigenemia
in the absence of detectable viremia.79 In-practice lateral flow
assays are available that detect antigen in anticoagulated whole
blood, plasma, or serum. In the past, the use of whole blood
generated less reliable results than when plasma or serum was
used, but with new-generation tests, whole blood is considered
an acceptable alternative.79 When a choice is available, serum is
FIGURE 22-6  Lateral thoracic radiograph from a 5-year-old male neutered domes-
tic shorthair cat with mediastinal lymphoma 11 months after it tested positive for cir- the preferred specimen. The use of tears or saliva is not recom-
culating FeLV antigen. The cat was tachypneic and had muffled heart sounds that were mended, because errors are more likely to occur. When virus
displaced caudally. There is evidence of pleural effusion and dorsal displacement of the isolation in culture was used as the gold standard, the sensitivity
trachea. Cytologic examination of pleural fluid revealed large numbers of malignant of seven different assays ranged from 92.1% to 96.8%, and the
lymphocytes. specificity ranged from 95.4% to 99.2%.81

TABLE 22-2
Diagnostic Assays Available for Feline Leukemia Virus Infection
Assay Specimen Type Target Performance
ELISA or ­similar Serum, plasma, whole FeLV p27 antigen Confirmation of positive results is recommended in healthy
immuno-­ blood cats with a second test from a different manufacturer.
chromatographic Positive antigen test results do not signify progressive
tests for soluble infection, and the assay must be repeated in 1 to 3 months
FeLV antigen or an IFA performed. False negatives can occur in the first
month of infection.
IFA Serum, bone marrow FeLV antigen in Less sensitive than ELISA. Positive results indicate infection
blood cells of the bone marrow and therefore progressive infection.
False positives may occur if nonspecific fluorescence is
interpreted as a positive result.
PCR Blood, bone marrow, FeLV RNA (RT-PCR) Sensitivity and specificity may vary between laboratories.
saliva (RT-PCR); or ­proviral DNA Never use in the absence of antigen testing. Assays that
bone marrow, tissue, have demonstrated sensitivity and specificity may be ­useful
lymph node aspirates to detect cats with regressive infection for ­elimination
(PCR) from blood donor programs, or to resolve the results
of discordant ELISA and IFA assays. False-negative test
results may occur when variant strains are present.
Virus isolation Blood, bone marrow Replication-competent Difficult, not widely available. Requires a specialized
FeLV virus ­laboratory. Used primarily as a research tool.

IFA, immunofluorescent antibody; RT, reverse transcriptase.


CHAPTER 22  Feline Leukemia Virus Infection 231

When ELISAs are used as screening tests, confirmation of there are inadequate blood cells in the periphery, such as in
positive test results is recommended because of the low preva- neutropenic cats. Performing IFA on bone marrow rather than
lence of infection in healthy cats and the higher possibility that peripheral blood may help to overcome this problem. False-pos-
false-positive test results may occur. Positive test results in the itive results can occur when inexperienced laboratory personnel
absence of FeLV antigen have the potential to occur rarely as a interpret nonspecific fluorescence as a positive test result. They
result of operator error or nonspecific reactivity.82 As for FIV also have the potential to occur when the antibody conjugates
infection, it is especially important to immediately confirm posi- bind nonspecifically to eosinophil granules, so eosinophils must
tive test results if they are likely to result in euthanasia or rehom- be excluded in interpretation of fluorescing cells.83
ing for disease control purposes. There are several options to Some cats with clinical abnormalities that strongly suggest
confirm a positive test result: FeLV-related disease (such as leukemia or myelodysplasia) test
• Perform another ELISA antigen test using an assay from a negative for circulating antigen using ELISA but have bone
different manufacturer. However, it should be remembered marrow cells that test positive for FeLV antigen using IFA. For
that in contrast to FIV infection, cats that test truly posi- example, at the author’s teaching hospital, 8 of 18 cats with
tive for FeLV antigen early in the course of infection (i.e., leukemia or MDS that tested negative for soluble FeLV anti-
before involvement of the bone marrow) may ultimately con- gen had bone marrow smears that were positive using IFA; the
trol the infection. Thus a single positive test result does not remainder tested IFA negative. This phenomenon may reflect
imply progressive infection, even if it is immediately repeat- either false-positive IFA assay results, or true infection with
able using a test from a different manufacturer. If the cat has undetectable levels of antigen in the peripheral blood. The use
signs consistent with FeLV-related disease, a single positive of PCR on bone marrow may help to resolve the FeLV status in
test result is more likely to mean that progressive infection is at least some of these cats.
present. Immunohistochemistry can also be used to detect viral anti-
• Perform an IFA assay on peripheral blood smears, because gen in tissue specimens or bone marrow core biopsies, although
cats with positive IFA results have infection of the bone mar- it may be less sensitive than IFA.
row and, with rare exceptions, are almost always progres-
sively infected. Cats that test negative with IFA assays may Molecular Diagnosis Using the Polymerase Chain Reaction
be in a transient viremic phase that may result in either pro- Several different PCR assays have been developed for detection
gressive or regressive infection, or they may have progressive of FeLV nucleic acid. Currently the major clinical indications
infection but the sensitivity of IFA is too low to detect it. In for PCR are (1) to screen potential blood donors in conjunction
this case, both ELISA and IFA assays, or the ELISA assay with antigen testing or (2) to test for regressive infection when
alone, could be repeated in 1 to 4 months. FeLV is strongly suspected as the cause of neoplasia but antigen
• Retest with ELISA 6 months later. If the antigen test remains tests are negative.
positive, progressive infection is likely. In some cats, antigen- PCR assays may detect FeLV RNA (reverse transcriptase–
emia persists for 16 weeks before regression occurs, so the polymerase chain reaction [RT-PCR]) or proviral DNA and
test could be repeated earlier than 6 months (e.g., 12 weeks must be carefully designed so that they do not detect endog-
later) or monthly if client finances permit so long as the cat enous FeLV sequences. At the current time, PCR assays should
remains healthy. never be used in the absence of antigen testing in order to screen
• Perform a full CBC. If hematologic abnormalities are pres- for or diagnose FeLV infection. The clinician needs to under-
ent, progressive infection is likely. stand if the assay used detects proviral DNA, viral RNA, or
Negative ELISA results can occur in the first month after expo- both (some laboratories run both assays), because the clinical
sure to FeLV, before sufficient antigen is detectable in the significance of a positive viral RNA assay (i.e., productive viral
peripheral blood. Cats that test negative within 30 days of pos- infection) differs from that of a positive proviral DNA assay
sible exposure to the virus should be retested 1 to 2 months (which suggests nonproductive viral infection for cats with neg-
later. Because development of antibodies to FIV can take up to 2 ative antigen tests). After infection, RT-PCR assays may be posi-
months, it is usually most practical to retest for both viral infec- tive several weeks before antigen tests or virus isolation become
tions 2 months after possible exposure. Kittens can be tested positive,84 and depending on the assay, PCR for viral RNA can
at any time, because maternal antibody does not interfere with be more sensitive than soluble antigen tests. High viral RNA
FeLV testing. loads in blood and saliva appear to be associated with progres-
sive infection, whereas low loads may be associated with regres-
Immunofluorescent Antibody or Immunoperoxidase Staining sive infection (Figure 22-7).84
IFA assays are widely offered by veterinary diagnostic labora- PCR assays for proviral DNA can be used on blood, buffy
tories and can be performed on fresh peripheral blood or bone coats, bone marrow, or tissues of cats that test negative for FeLV
marrow. At least two fresh smears (without anticoagulant) antigen. Cats with a positive proviral PCR test result but nega-
should be air-dried and mailed to the laboratory. IFA is less tive soluble antigen test, which in one study represented about
sensitive than ELISA and, depending on the laboratory, is more 10% of cats with negative antigen test results, have regressive
prone to false-negative and false-positive results and so is not infection.85,86 These cats are probably not infectious to other cats
recommended for screening purposes. The presence of detect- but may reactivate virus shedding with severe stress or immu-
able virus using IFA in circulating blood cells indicates pro- nosuppression, or transmit the virus through blood transfusion
gressive infection more than 90% of the time. Cats with early or vertical transmission. Proviral DNA appears to be present
viremia (before the bone marrow is infected) test IFA-negative at much lower levels in cats with regressive infection than in
but ELISA-positive. Cats with regressive infection test negative those with progressive infection.86,87 The cell types that contain
with both IFA and ELISA assays (see Figure 22-2). Negative IFA FeLV proviral DNA also may differ for antigen-negative and
test results can occur in cats with progressive infection when antigen-positive cats. The proviral DNA of one FeLV strain was
232 SECTION 1  Viral Diseases

A B C D

E F G H
FIGURE 22-7  Viral RNA (A-D) and p27 antigen (E-H) loads in plasma (A and E), saliva (B and F), feces (C and G), and urine (D and H) in 10 cats experimentally infected with FeLV.
Each line represents observations for a single cat. Continuous lines represent data from cats with progressive infection, and broken lines represent data from cats with regressive infection.
(Modified from Cattori V, Randon R, Riond B, et al. The kinetics of feline leukemia virus shedding in experimentally infected cats are associated with outcome. Vet Microbiol 2009;133(3):292-
296, Figure 1.)

found only in the lymphocytes of antigen-negative cats, whereas usually fail to develop antibody responses.24,48,88 Antibody
antigen-positive cats had high loads within all leukocyte types, responses may occur after vaccination with FeLV vaccines.
including lymphocytes, monocytes, and granulocytes.86,87 The
use of bone marrow or tissue specimens, rather than whole Pathologic Findings
blood, may increase sensitivity for detection of proviral DNA. Gross and histopathologic findings in cats with FeLV infection
The sensitivity and specificity of commercially available usually reflect secondary disease processes. Lymphoma and evi-
PCR assays are likely to vary with assay design, and the useful- dence of myelodysplasia or leukemias are the most common
ness of assays offered commercially has not been published, so findings, but opportunistic infections may also be detected.
until more information is available, great caution is warranted The intestinal tracts of cats with FeLV-associated enteritis
when interpreting the results of PCR assays for FeLV infection. reveal crypt cell necrosis and regeneration, lymphoplasmacytic
Retroviruses have high rates of mutation, and the presence of infiltrates, and blunting and fusion of villi (See Figure 22-5).90
sequence variations may lead to false-negative results. False- Reactive lymphoid hyperplasia is also a common finding. CNS
positive results occur in some PCR laboratories as a result of lesions include loss of axons and dilated myelin sheaths within
contamination or manual loading errors, or poor assay design. the spinal cord.74 Immunohistochemical stains may be applied
Because FeLV vaccine virus is inactivated or recombinant, it to tissue sections to confirm the presence of FeLV antigen in
does not replicate or integrate into the host genome, so vaccina- association with lesions.
tion should not lead to false-positive PCR results.
Treatment and Prognosis
Virus Isolation
FeLV can be readily isolated in cell culture. The cell culture Cats with opportunistic infections and lymphoma can be suc-
supernatant can then be tested for production of FeLV using cessfully treated using the same medications and supportive
antigen assays or viral RNA PCR. Because growth requires sev- treatments used for FeLV negative cats with these problems.
eral days and specialized techniques, cell culture is not routinely Opportunistic infections may require longer periods of treat-
used for clinical diagnosis. ment or, in some cases, lifelong treatment with antimicrobial
drugs. Hemoplasma infections should be treated with doxycy-
Antibody Assays cline (see Chapter 41). Cats with nonregenerative anemias may
Although not useful for diagnosis of FeLV infection, detec- require periodic blood transfusions. Serum erythropoietin con-
tion of antibody responses to FeLV infection is performed on a centrations are already high in these cats, and in many cases,
clinical research basis with virus neutralization assays and IFA treatment with darbepoetin or human recombinant erythro-
assays that detect antibodies against feline oncornavirus cell poietin is unsuccessful but could be attempted. The efficacy of
membrane–associated antigen (FOCMA).24,48,84,88,89 Positive treatments such as filgrastim is not known and may be com-
antibody test results in cats that test negative for FeLV antigen plicated by the formation of antibodies (see Chapter 7). The
indicate previous exposure followed by abortive or regressive management of cats with stomatitis is discussed in Chapter 21.
infection. Cats with progressive infections and high viral loads In general, glucocorticoids and other immunosuppressive drugs
CHAPTER 22  Feline Leukemia Virus Infection 233

TABLE 22-3
Suggested Medications for Treatment of Cats with Feline Leukemia Virus Infection
Drug Dose Route Interval (hours) Comments
Zidovudine (AZT) 5 mg/kg PO 12 Monitor CBC weekly
during treatment for
the first month, then
monthly.
Feline recombinant interferon omega 1 million U/kg SC q24h for 5 consecutive days
­starting on days 0, 14, and 60
Human recombinant interferon alpha 1 to 50 U/cat PO 24

should be avoided unless immune-mediated cytopenias are sus- Some cats with FeLV-associated lymphoma may have long-term
pected. Some cats with FeLV-associated IMHA respond well to remissions when treated with standard chemotherapy protocols.
treatment with glucocorticoids, and glucocorticoid treatment Some, but not all, studies suggest that FeLV infection is a nega-
may be unavoidable. tive prognostic indicator in cats with lymphoma.96 One of the
Antiviral agents and immunomodulators are of limited ben- authors has treated a cat that initially tested positive for FeLV
efit for treatment of cats with FeLV infections (see Chapter 7 antigen using ELISA and had evidence of IFA-positive, severe
for more information) (Table 22-3). The use of feline recom- megaloblastic erythroid dysplasia in the marrow and IMHA, yet
binant IFN-ω improved clinical scores and survival times in subsequently became ELISA-negative and remained in clinical
cats with FeLV infection over a short time period (2 months) in remission while being treated with glucocorticoids more than
one study.91 Beneficial outcomes have also been described after 4 years later.97 The prognosis is least favorable for cats with
treatment with low-dose oral human recombinant IFN-α. Zid- leukemia, which generally survive less than a few weeks.
ovudine (AZT) has not performed as well for treatment of sick
cats with FeLV infections as it has for those with FIV infections, Immunity and Vaccination
and the results of some studies have shown minimal benefit. In
other studies, AZT improved oral cavity inflammation, reduced Several parenteral vaccines are available for prevention of FeLV
antigenemia, and prolonged life span in naturally and experi- infection, which include adjuvanted inactivated whole virus
mentally infected cats with FeLV infection.92,93 Controlled stud- vaccines; nonadjuvanted canarypox vectored virus vaccines
ies of the efficacy of other treatments, such as lymphocyte T-cell that incorporate the env and gag genes of FeLV (Purevax and
immunomodulator (T-cyte Therapeutics, Inc.) and acemannan, Eurifel, Merial); and a recombinant subunit vaccine that con-
are required. Antivirals that show promise for treatment of tains p45, the nonglycosylated form of gp70 (Leucogen, Vir-
FeLV infection include fozivudine (which is closely related to bac). The recombinant canarypox vaccine available in Europe
AZT) and the integrase inhibitor raltegravir, which has been is a different product than that licensed for use in the United
used to treat gammaretrovirus infections in humans and inhibits States. Studies that have used highly sensitive PCR assays have
FeLV replication in cell culture.94 The safety of these drugs in shown that vaccination does not produce sterilizing immunity
cats remains to be determined. (that is, complete absence of viral RNA, DNA, antigenemia, and
Cats infected with FeLV should be housed indoors to prevent viremia after challenge).98 In other words, cats develop regres-
spread of infection to other cats. Indoor housing also minimizes sive or even progressive infections, but not abortive infections
exposure of infected cats to other opportunistic pathogens. after challenge.98 Nevertheless, vaccination with recombinant
Raw food diets should not be fed. Survival may be prolonged subunit, canarypox vectored, and whole virus vaccine prevented
in low-stress environments, so provision of space, adequate lit- progressive infection in 87%, 78%, and 44% of cats when com-
ter boxes, management of co-infections, and a proper diet are pared with controls, respectively. One vaccine protected 83% of
important. Vaccines administered for prevention of respiratory cats against antigenemia after challenge as long as 2 years after
viruses and FPV should be inactivated. Some FeLV-infected cats vaccination.99
may not respond as well to vaccination as noninfected cats. In summary, no vaccine provides 100% protection against
FeLV infection, and even when protection against progressive
Prognosis infection occurs, regressive infections still occur. However,
Survival times vary considerably depending on the stage of because vaccination protects cats from progressive infection, it
infection, host immunity, and the strain of FeLV involved. Nev- is indicated for all at-risk cats, such as those with outdoor expo-
ertheless, virtually all cats that are progressively infected with sure or those that reside in households with other FeLV antigen-
FeLV go on to develop FeLV-related disease within 5 years of positive cats. The American Association of Feline Practitioners
diagnosis.95 A comparison of more than 800 FeLV-infected cats (AAFP) highly recommends that all FeLV antigen-negative kit-
and 8000 controls revealed a median survival time of 2.4 years tens be vaccinated for FeLV, because of the potential that some
for FeLV-infected cats versus 6.3 years for controls.79 Many of these kittens may escape or become outdoor cats, even when
progressively infected cats, especially adult cats, may live for the intention of the owner is to keep the kitten indoors.100 Vac-
several years with a good quality of life, and so euthanasia is cination is also recommended for cats entering shelters that are
not recommended on the basis of a positive FeLV test alone. likely to be housed with other cats.79 Two doses are given in
234 SECTION 1  Viral Diseases

the left pelvic limb as distally as possible, 3 to 4 weeks apart cats, vaccination, and proper screening of blood donor cats
from 8 to 9 weeks of age, followed by a booster at 1 year and with soluble antigen assays and PCR. Routine hand-washing
then every 1 to 3 years thereafter, although more information precautions are indicated for hospitalized cats, and precau-
is required on the duration of immunity for FeLV vaccines. The tions should be taken (such as the wearing of gloves) to protect
European Advisory Bureau on Cat Diseases (ABCD) suggests a FeLV-infected cats from nosocomial pathogens. Fomites such
booster every 2 to 3 years for cats older than 3 to 4 years of age, as food and water bowls and litter boxes should not be shared
in light of the lower susceptibility of adult cats to infection.101 between FeLV antigen-negative cats and cats with unknown or
Annual boosters are required for recombinant vaccines. Acutely antigen-positive FeLV status. Neutering can prevent roaming
ill cats should not be vaccinated, but it is acceptable to vacci- and reduce the likelihood of FeLV infection. When a cat from
nate cats with chronic diseases, such as chronic kidney disease. a multicat household tests antigen-positive, all cats should be
Testing for FeLV should be performed before each booster if tested and retested and positive cats should be separated from
exposure to FeLV was likely before the booster was required other cats if possible. Euthanasia is recommended for sick,
(which should apply for most cats vaccinated for FeLV). Vac- FeLV-positive cats that enter shelters.101 People who adopt cats
cination with FeLV vaccines has been associated with injection- that have unknown retrovirus status from shelters should be
site sarcomas, so only cats that are likely to be exposed should educated about the disease and the need for quarantine and test-
be vaccinated. ing after adoption.

Prevention Public Health Aspects


Prevention of FeLV infection involves indoor housing of cats Although FeLV can replicate in human cell culture lines, no con-
away from other cats infected with FeLV, testing and removal clusive evidence of natural infection with FeLV has ever been
(or separation) of cats that test antigen-positive from other detected in humans.

CASE EXAMPLE Cardiovascular and Respiratory: No clinically significant


abnormalities were detected.
Gastrointestinal and Urogenital: The cat resented palpation
Signalment: “Hamster” a 6-year-old female spayed domestic of her cranial abdomen. The perianal region was stained
shorthair from Fairfield, CA. with green fecal material.
History: Hamster was brought to the University of California, Lymph Nodes: All lymph nodes were <1 cm in diameter.
Davis, Veterinary Medical Teaching Hospital for the Laboratory Findings:
problems of fever, lethargy, anorexia, diarrhea, and CBC:
vomiting. The diarrhea had been present for 3 days and HCT 24.6% (30%-50%).
was greenish, mucoid, and occurred once to twice daily. MCV 48.4 fL (42-53 fL), MCHC 32.1 g/dL (30-33.5 g/dL)
The owner reported that Hamster had vomited yellow fluid Reticulocytes 7800 cells/µL
at least three times during this period and was completely WBC 11,470 cells/µL (4500-14,000 cells/µL)
inappetent. Her urination habits had not changed. Hamster Neutrophils 10,438 cells/µL (2000-9000 cells/µL)
was taken to a local veterinary clinic on the first day of illness, Band neutrophils 459 cells/µL
where a physical examination revealed pyrexia (105.1°F or Lymphocytes 344 cells/µL (1000-7000 cells/µL)
40.6°C). She was treated with clavulanic acid–amoxicillin Monocytes 229 cells/µL (50-600 cells/µL)
and a single intramuscular injection of enrofloxacin but Platelets 122,000 platelets/µL (180,000-500,000 platelets/
there was no clinical improvement. Hamster had tested µL). The neutrophils showed slight toxicity, and there
positive for FeLV antigen 1.5 years ago, after which time she were a few macroplatelets
had been housed exclusively indoors. She lived with some Serum Chemistry Profile:
caged birds and three other FeLV-positive cats. None of Sodium 148 mmol/L (151-158 mmol/L)
the other cats were ill. Hamster played with elastic bands, Potassium 3.8 mmol/L (3.6-4.9 mmol/L)
but there was no known toxin exposure and no recent Chloride 110 mmol/L (117-126 mmol/L)
changes in her environment. Her diet usually consisted of a Bicarbonate 18 mmol/L (15-21 mmol/L)
commercial dry cat food. Phosphorus 2.9 mg/dL (3.2-6.3 mg/dL)
Physical Examination: Calcium 9.5 mg/dL (9.0-10.9 mg/dL)
Body Weight: 6.9 kg BUN 15 mg/dL (18-33 mg/dL)
General: Quiet, alert, responsive, hydrated, T = 104.4°F Creatinine 1.2 mg/dL (1.1-2.2 mg/dL)
(40.2°C), HR = 220 beats/min, RR = 36 breaths/min, mucous Glucose 169 mg/dL (63-118 mg/dL)
membranes pink, CRT = 1 s. Haircoat unkempt. Total protein 7.0 g/dL (6.6-8.4 g/dL)
Eyes, Ears, Nose, and Throat (with Dilated Fundoscopic Albumin 2.6 g/dL (2.2-4.6 g/dL)
Examination): The only abnormality noted was mild Globulin 4.4 g/dL (2.8-5.4 g/dL)
gingivitis. ALT 72 U/L (27-101 U/L), AST 121 U/L (17-58 U/L)
Musculoskeletal: Body condition score 7/9. Normal ALP 12 U/L (14-71 U/L), γ- GGT 0 U/L (0-4 U/L)
ambulation was present. Cholesterol 104 mg/dL (89-258 mg/dL)
Total bilirubin 0.2 mg/dL (0-0.2 mg/dL).
CHAPTER 22  Feline Leukemia Virus Infection 235

Urinalysis (Cystocentesis): SGr 1.020; pH 7.0, 1+ protein, 2+ physical examination, CBC, kidney panel, urinalysis, and
hemoprotein, 0-1 WBC/HPF, 0-2 RBC/HPF, rare rods, few aerobic bacterial urine culture showed no abnormalities.
amorphous crystals. Her hematocrit was 38.9%, white cell count was 4620 cells/
Imaging Findings: µL, and platelet count was 220,000/µL. One year later, she
Plain Abdominal Radiographs: The small bowel was diffusely remained clinically healthy and a CBC showed a hematocrit
gas- and fluid-filled with normal intestinal diameter. The of 38.2%, white cell count of 4500 cells/µL, neutrophil count
colon was relatively empty with a small amount of fluid. No of 3362 cells/µL, lymphocyte count of 905 cells/µL, and a
masses were identified. There was renal asymmetry with monocyte count of 45 cells/µL. Despite the mild cytopenias
poor visualization of the right kidney. Abdominal serosal present, Hamster remained alive and well with a normal CBC
detail appeared normal. 4 years after she was initially seen for gastrointestinal signs.
Abdominal Sonography: The right kidney could not be She died 2 years after that, 7.5 years after she first tested
identified. The left kidney appeared normal and measured positive for FeLV infection.
4.2 cm in length. A small amount of fluid was present in the One of the other cats in the household, a 5-year-old, male neu-
colon. The remainder of the abdomen was unremarkable. tered domestic medium hair cat, was euthanized 1.5 years
Microbiologic and Virologic Testing: Aerobic bacterial after Hamster was initially evaluated. He developed severe
urine culture: negative normocytic, normochromic nonregenerative anemia (HCT
Point-of-care ELISA serology for FeLV antigen and FIV antibody: 10.5%, with 5200 reticulocytes and 11 nucleated RBC/HPF)
positive for FeLV antigen with neutropenia (1320 cells/µL) in association with circulat-
PCR for Mycoplasma haemofelis and Candidatus Mycoplasma ing blast cells.
haemominutum (whole blood): negative Comments: Even though this cat was initially seen for
Diagnosis: Progressive FeLV infection with suspected acute moderately severe illness in association with progressive
pyelonephritis (characterized by fever, gastrointestinal FeLV infection, she made a full recovery with treatment
signs, bacteriuria, neutrophilia with a left shift, and mild and remained alive and well for 6 additional years. The
thrombocytopenia). Right renal agenesis (likely congenital). nonregenerative anemia likely resulted from inflammatory
Treatment: Hamster was hospitalized and treated with disease. The negative urine culture may have resulted
intravenous fluids, enrofloxacin (5 mg/kg, slow IV, q24h) and from recent treatment with antimicrobial drugs, and renal
ampicillin (20 mg/kg, IV, q8h) (note that the parenteral use of ultrasound is insensitive for diagnosis of pyelonephritis.
enrofloxacin in this cat was off-label and has the potential to The age of this cat, the strain of FeLV involved, and the
cause irreversible blindness). Her temperature normalized challenge dose, as well as other environmental and genetic
within 24 hours, and there was no more vomiting or diarrhea. factors, probably all played a role in the course of disease.
After 48 hours, the hematocrit was 27.6%, neutrophil This case clearly demonstrates that other treatable diseases
count was 5759 cells/µL with no bands or toxicity, and may be present in FeLV-infected cats and that search for the
lymphocyte count was 917 cells/µL. She was discharged underlying cause of clinical signs and adequate treatment
from the hospital with instructions to continue antibiotic is always recommended. The second cat in the household
treatment for 1 month. Two weeks after the antibiotics likely also had progressive FeLV infection; the development
had been discontinued, she was apparently healthy and a of anemia suggested infection with FeLV-C.

SUGGESTED READINGS 4. Lauring AS, Anderson MM, Overbaugh J. Specificity in receptor


usage by T-cell-tropic feline leukemia viruses: implications for the
Hartmann K. Clinical aspects of feline immunodeficiency and feline in  vivo tropism of immunodeficiency-inducing variants. J Virol.
leukemia virus infection. Vet Immunol Immunopathol. 2011;143: 2001;75:8888-8898.
190-201. 5. Brojatsch J, Kristal BS, Viglianti GA, et al. Feline leukemia virus
Levy J, Crawford C, Hartmann K, et al. 2008 American Association subgroup C phenotype evolves through distinct alterations near
of Feline Practitioners’ feline retrovirus management guidelines. J the N terminus of the envelope surface glycoprotein. Proc Natl
Feline Med Surg. 2008;10:300-316. Acad Sci U S A. 1992;89:8457-8461.
Lutz H, Addie D, Belak S, et al. Feline leukaemia. ABCD guidelines on 6. Chandhasin C, Coan PN, Levy LS. Subtle mutational changes in
prevention and management. J Feline Med Surg. 2009;11:565-574. the SU protein of a natural feline leukemia virus subgroup A iso-
late alter disease spectrum. J Virol. 2005;79:1351-1360.
REFERENCES 7. Chandhasin C, Coan PN, Pandrea I, et  al. Unique long termi-
1. Jarrett WFH, Crawford EM, Martin WB, et  al. A virus-like nal repeat and surface glycoprotein gene sequences of feline
particle associated with leukemia (lymphosarcoma). Nature. leukemia virus as determinants of disease outcome. J Virol.
1964;202:567-569. 2005;79:5278-5287.
2. Meli ML, Cattori V, Martinez F, et  al. Feline leukemia virus 8. Vobis M, D’Haese J, Mehlhorn H, et al. Evidence of horizontal
and other pathogens as important threats to the survival of the transmission of feline leukemia virus by the cat flea (Ctenocephali-
critically endangered Iberian lynx (Lynx pardinus). PLoS One. des felis). Parasitol Res. 2003;91:467-470.
2009;4:e4744. 9. Gomes-Keller MA, Gonczi E, Grenacher B, et al. Fecal shedding
3. Stewart MA, Warnock M, Wheeler A, et al. Nucleotide sequences of infectious feline leukemia virus and its nucleic acids: a transmis-
of a feline leukemia virus subgroup A envelope gene and long ter- sion potential. Vet Microbiol. 2009;134:208-217.
minal repeat and evidence for the recombinational origin of sub- 10. Pacitti AM, Jarrett O, Hay D. Transmission of feline leukaemia
group B viruses. J Virol. 1986;58:825-834. virus in the milk of a non-viraemic cat. Vet Rec. 1986;118:381-384.
236 SECTION 1  Viral Diseases

11. Tenorio AP, Franti CE, Madewell BR, et al. Chronic oral infec- 31. Mathes LE, Olsen RG, Hebebrand LC, et al. Abrogation of lym-
tions of cats and their relationship to persistent oral carriage of phocyte blastogenesis by a feline leukaemia virus protein. Nature.
feline calici-, immunodeficiency, or leukemia viruses. Vet Immu- 1978;274:687-689.
nol Immunopathol. 1991;29:1-14. 32. Perryman LE, Hoover EA, Yohn DS. Immunologic reactivity of
12. O’Connor Jr TP, Tonelli QJ, Scarlett JM. Report of the the cat: immunosuppression in experimental feline leukemia.
National FeLV/FIV Awareness Project. J Am Vet Med Assoc. J Natl Cancer Inst. 1972;49:1357-1365.
1991;199:1348-1353. 33. Trainin Z, Wernicke D, Ungar-Waron H, et al. Suppression of the
13. Levy JK, Scott HM, Lachtara JL, et  al. Seroprevalence of feline humoral antibody response in natural retrovirus infections. Sci-
leukemia virus and feline immunodeficiency virus infection among ence. 1983;220:858-859.
cats in North America and risk factors for seropositivity. J Am Vet 34. Wernicke D, Trainin Z, Ungar-Waron H, et al. Humoral immune
Med Assoc. 2006;228:371-376. response of asymptomatic cats naturally infected with feline leu-
14. Stutzer B, Simon K, Lutz H, et al. Incidence of persistent viraemia kemia virus. J Virol. 1986;60:669-673.
and latent feline leukaemia virus infection in cats with lymphoma. 35. Lafrado LJ, Olsen RG. Demonstration of depressed polymorpho-
J Feline Med Surg. 2011;13:81-87. nuclear leukocyte function in nonviremic FeLV-infected cats. Can-
15. Gleich SE, Krieger S, Hartmann K. Prevalence of feline immuno- cer Invest. 1986;4:297-300.
deficiency virus and feline leukaemia virus among client-owned 36. Hoffmann-Jagielska M, Winnicka A, Jagielski D, et al. Influence
cats and risk factors for infection in Germany. J Feline Med Surg. of naturally acquired feline leukemia virus (FeLV) infection on
2009;11:985-992. the phagocytic and respiratory burst activity of neutrophils and
16. Ravi M, Wobeser GA, Taylor SM, et al. Naturally acquired feline monocytes of peripheral blood. Pol J Vet Sci. 2005;8:93-97.
immunodeficiency virus (FIV) infection in cats from western Can- 37. Wardini AB, Guimaraes-Costa AB, Nascimento MT, et al. Char-
ada: Prevalence, disease associations, and survival analysis. Can acterization of neutrophil extracellular traps in cats naturally
Vet J. 2010;51:271-276. infected with feline leukemia virus. J Gen Virol. 2010;91:259-264.
17. Little S, Sears W, Lachtara J, et al. Seroprevalence of feline leuke- 38. Buchmann AU, Kershaw O, Kempf VA, et al. Does a feline leuke-
mia virus and feline immunodeficiency virus infection among cats mia virus infection pave the way for Bartonella henselae infection
in Canada. Can Vet J. 2009;50:644-648. in cats?. J Clin Microbiol. 2010;48:3295-3300.
18. Hellard E, Fouchet D, Santin-Janin H, et al. When cats’ ways of 39. Fujino Y, Ohno K, Tsujimoto H. Molecular pathogenesis of feline
life interact with their viruses: a study in 15 natural populations leukemia virus-induced malignancies: insertional mutagenesis. Vet
of owned and unowned cats (Felis silvestris catus). Prev Vet Med. Immunol Immunopathol. 2008;123:138-143.
2011;101:250-264. 40. Shelton GH, Grant CK, Cotter SM, et al. Feline immunodeficiency
19. Al-Kappany YM, Lappin MR, Kwok OC, et  al. Seropreva- virus and feline leukemia virus infections and their relationships to
lence of Toxoplasma gondii and concurrent Bartonella spp., lymphoid malignancies in cats: a retrospective study (1968-1988).
feline immunodeficiency virus, feline leukemia virus, and Diro- J Acquir Immune Defic Syndr. 1990;3:623-630.
filaria immitis infections in Egyptian cats. J Parasitol. 2011;97: 41. Reinacher M. Diseases associated with spontaneous feline leuke-
256-258. mia virus (FeLV) infection in cats. Vet Immunol Immunopathol.
20. Hosie MJ, Robertson C, Jarrett O. Prevalence of feline leukaemia 1989;21:85-95.
virus and antibodies to feline immunodeficiency virus in cats in the 42. Louwerens M, London CA, Pedersen NC, et al. Feline lymphoma in
United Kingdom. Vet Rec. 1989;125:293-297. the post-feline leukemia virus era. J Vet Intern Med. 2005;19:329-335.
21. Grant CK, Essex M, Gardner MB, et al. Natural feline leukemia 43. Teske E, van Straten G, van Noort R, et al. Chemotherapy with
virus infection and the immune response of cats of different ages. cyclophosphamide, vincristine, and prednisolone (COP) in cats
Cancer Res. 1980;40:823-829. with malignant lymphoma: new results with an old protocol.
22. Hofmann-Lehmann R, Tandon R, Boretti FS, et al. Reassessment J Vet Intern Med. 2002;16:179-186.
of feline leukaemia virus (FeLV) vaccines with novel sensitive 44. Taylor SS, Goodfellow MR, Browne WJ, et al. Feline extranodal
molecular assays. Vaccine. 2006;24:1087-1094. lymphoma: response to chemotherapy and survival in 110 cats.
23. Kadar E, Sykes JE, Kass PH, et al. Evaluation of the prevalence J Small Anim Pract. 2009;50:584-592.
of infections in cats after renal transplantation: 169 cases (1987- 45. Krick EL, Little L, Patel R, et  al. Description of clinical and
2003). J Am Vet Med Assoc. 2005;227:948-953. pathological findings, treatment and outcome of feline large
24. Major A, Cattori V, Boenzli E, et al. Exposure of cats to low doses granular lymphocyte lymphoma (1996-2004). Vet Comp Oncol.
of FeLV: seroconversion as the sole parameter of infection. Vet 2008;6:102-110.
Res. 2010;41:17. 46. Francis DP, Essex M, Cotter SM, et al. Epidemiologic association
25. Good RA, Ogasawara M, Liu WT, et  al. Immunosuppressive between virus-negative feline leukemia and the horizontally trans-
actions of retroviruses. Lymphology. 1990;23:56-59. mitted feline leukemia virus. Cancer Lett. 1981;12:37-42.
26. Cianciolo GJ, Copeland TD, Oroszlan S, et al. Inhibition of lym- 47. Wang J, Kyaw-Tanner M, Lee C, et al. Characterisation of lympho-
phocyte proliferation by a synthetic peptide homologous to retro- sarcomas in Australian cats using polymerase chain reaction and
viral envelope proteins. Science. 1985;230:453-455. immunohistochemical examination. Aust Vet J. 2001;79:41-46.
27. Mitani M, Cianciolo GJ, Snyderman R, et al. Suppressive effect on 48. Beatty JA, Tasker S, Jarrett O, et al. Markers of feline leukaemia
polyclonal B-cell activation of a synthetic peptide homologous to virus infection or exposure in cats from a region of low seropreva-
a transmembrane component of oncogenic retroviruses. Proc Natl lence. J Feline Med Surg. 2011;13:927-933.
Acad Sci U S A. 1987;84:237-240. 49. Jackson ML, Haines DM, Meric SM, et al. Feline leukemia virus
28. Haraguchi S, Good RA, Day-Good NK. A potent immunosup- detection by immunohistochemistry and polymerase chain reac-
pressive retroviral peptide: cytokine patterns and signaling path- tion in formalin-fixed, paraffin-embedded tumor tissue from cats
ways. Immunol Res. 2008;41:46-55. with lymphosarcoma. Can J Vet Res. 1993;57:269-276.
29. Cockerell GL, Hoover EA, Krakowka S, et al. Lymphocyte mito- 50. Gabor LJ, Jackson ML, Trask B, et al. Feline leukaemia virus status of
gen reactivity and enumeration of circulating B- and T-cells dur- Australian cats with lymphosarcoma. Aust Vet J. 2001;79:476-481.
ing feline leukemia virus infection in the cat. J Natl Cancer Inst. 51. Gelain ME, Antoniazzi E, Bertazzolo W, et  al. Chronic eosino-
1976;57:1095-1099. philic leukemia in a cat: cytochemical and immunophenotypical
30. Hebebrand LC, Mathes LE, Olsen RG. Inhibition of concanavalin features. Vet Clin Pathol. 2006;35:454-459.
A stimulation of feline lymphocytes by inactivated feline leukemia 52. Shimoda T, Shiranaga N, Mashita T, et al. Chronic myelomono-
virus. Cancer Res. 1977;37:4532-4533. cytic leukemia in a cat. J Vet Med Sci. 2000;62:195-197.
CHAPTER 22  Feline Leukemia Virus Infection 237

53. Besmer P, Lader E, George PC, et  al. A new acute transform- 73. Mitchell TW, Rojko JL, Hartke JR, et al. FeLV envelope protein
ing feline retrovirus with fms homology specifies a C-termi- (gp70) variable region 5 causes alterations in calcium homeostasis
nally truncated version of the c-fms protein that is different and toxicity of neurons. J Acquir Immune Defic Syndr Hum Ret-
from SM-feline sarcoma virus v-fms protein. J Virol. 1986;60: rovirol. 1997;14:307-320.
194-203. 74. Carmichael KP, Bienzle D, McDonnell JJ. Feline leukemia virus-
54. Donner L, Fedele LA, Garon CF, et  al. McDonough feline sar- associated myelopathy in cats. Vet Pathol. 2002;39:536-545.
coma virus: characterization of the molecularly cloned provirus 75. Kipar A, Kremendahl J, Jackson ML, et al. Comparative examina-
and its feline oncogene (v-fms). J Virol. 1982;41:489-500. tion of cats with feline leukemia virus-associated enteritis and other
55. Stiles J, Bienzle D, Render JA, et  al. Use of nested polymerase relevant forms of feline enteritis. Vet Pathol. 2001;38:359-371.
chain reaction (PCR) for detection of retroviruses from formalin- 76. Hardy Jr WD, Hess PW, MacEwen EG, et al. Biology of feline leuke-
fixed, paraffin-embedded uveal melanomas in cats. Vet Ophthal- mia virus in the natural environment. Cancer Res. 1976;36:582-588.
mol. 1999;2:113-116. 77. Lutz H, Castelli I, Ehrensperger F, et al. Panleukopenia-like syn-
56. Cullen CL, Haines DM, Jackson ML, et al. The use of immuno- drome of FeLV caused by co-infection with FeLV and feline pan-
histochemistry and the polymerase chain reaction for detection of leukopenia virus. Vet Immunol Immunopathol. 1995;46:21-33.
feline leukemia virus and feline sarcoma virus in six cases of feline 78. Kipar A, Kremendahl J, Addie DD, et al. Fatal enteritis associated
ocular sarcoma. Vet Ophthalmol. 1998;1:189-193. with coronavirus infection in cats. J Comp Pathol. 1998;119:1-14.
57. Cullen CL, Haines DM, Jackson ML, et al. Lack of detection of 79. Levy J, Crawford C, Hartmann K, et al. 2008 American Associa-
feline leukemia and feline sarcoma viruses in diffuse iris melano- tion of Feline Practitioners’ feline retrovirus management guide-
mas of cats by immunohistochemistry and polymerase chain reac- lines. J Feline Med Surg. 2008;10:300-316.
tion. J Vet Diagn Invest. 2002;14:340-343. 80. Gleich S, Hartmann K. Hematology and serum biochemistry of
58. Ellis JA, Jackson ML, Bartsch RC, et  al. Use of immunohisto- feline immunodeficiency virus-infected and feline leukemia virus-
chemistry and polymerase chain reaction for detection of oncor- infected cats. J Vet Intern Med. 2009;23:552-558.
naviruses in formalin-fixed, paraffin-embedded fibrosarcomas 81. Hartmann K, Griessmayr P, Schulz B, et al. Quality of different
from cats. J Am Vet Med Assoc. 1996;209:767-771. in-clinic test systems for feline immunodeficiency virus and feline
59. Kidney BA, Ellis JA, Haines DM, et  al. Comparison of endog- leukaemia virus infection. J Feline Med Surg. 2007;9:439-445.
enous feline leukemia virus RNA content in feline vaccine and 82. Barr MC, Pough MB, Jacobson RH, et al. Comparison and inter-
nonvaccine site-associated sarcomas. Am J Vet Res. 2001;62: pretation of diagnostic tests for feline immunodeficiency virus
1990-1994. infection. J Am Vet Med Assoc. 1991;199:1377-1381.
60. Hartmann K. Clinical aspects of feline immunodeficiency and 83. Floyd K, Suter PF, Lutz H. Granules of blood eosinophils are
feline leukemia virus infection. Vet Immunol Immunopathol. stained directly by anti-immunoglobulin fluorescein isothiocya-
2011;143:190-201. nate conjugates. Am J Vet Res. 1983;44:2060-2063.
61. Shelton GH, Linenberger ML. Hematologic abnormalities asso- 84. Cattori V, Tandon R, Riond B, et al. The kinetics of feline leukae-
ciated with retroviral infections in the cat. Semin Vet Med Surg mia virus shedding in experimentally infected cats are associated
(Small Anim). 1995;10:220-233. with infection outcome. Vet Microbiol. 2009;133:292-296.
62. Quigley JG, Yang Z, Worthington MT, et al. Identification of a 85. Gomes-Keller MA, Gonczi E, Tandon R, et al. Detection of feline
human heme exporter that is essential for erythropoiesis. Cell. leukemia virus RNA in saliva from naturally infected cats and cor-
2004;118:757-766. relation of PCR results with those of current diagnostic methods.
63. Hisasue M, Okayama H, Okayama T, et al. Hematologic abnor- J Clin Microbiol. 2006;44:916-922.
malities and outcome of 16 cats with myelodysplastic syndromes. 86. Hofmann-Lehmann R, Huder JB, Gruber S, et al. Feline leukae-
J Vet Intern Med. 2001;15:471-477. mia provirus load during the course of experimental infection and
64. Weiss DJ. Evaluation of dysmyelopoiesis in cats: 34 cases (1996- in naturally infected cats. J Gen Virol. 2001;82:1589-1596.
2005). J Am Vet Med Assoc. 2006;228:893-897. 87. Pepin AC, Tandon R, Cattori V, et  al. Cellular segregation of
65. Stutzer B, Muller F, Majzoub M, et al. Role of latent feline leu- feline leukemia provirus and viral RNA in leukocyte subsets of
kemia virus infection in nonregenerative cytopenias of cats. J Vet long-term experimentally infected cats. Virus Res. 2007;127:9-16.
Intern Med. 2010;24:192-197. 88. Harbour DA, Gunn-Moore DA, Gruffydd-Jones TJ, et al. Protec-
66. Anderson LJ, Jarrett WF. Membranous glomerulonephritis associ- tion against oronasal challenge with virulent feline leukaemia virus
ated with leukaemia in cats. Res Vet Sci. 1971;12:179-180. lasts for at least 12 months following a primary course of immuni-
67. Brightman 2nd AH, Ogilvie GK, Tompkins M. Ocular disease in sation with Leukocell 2 vaccine. Vaccine. 2002;20:2866-2872.
FeLV-positive cats: 11 cases (1981-1986). J Am Vet Med Assoc. 89. Torres AN, O’Halloran KP, Larson LJ, et al. Feline leukemia virus
1991;198:1049-1051. immunity induced by whole inactivated virus vaccination. Vet
68. Pedersen NC, Pool RR, O’Brien T. Feline chronic progressive Immunol Immunopathol. 2010;134:122-131.
polyarthritis. Am J Vet Res. 1980;41:522-535. 90. Reinacher M. Feline leukemia virus-associated enteritis—a condi-
69. Tuomari DL, Olsen RG, Singh VK, et  al. Detection of circulat- tion with features of feline panleukopenia. Vet Pathol. 1987;24:1-4.
ing immune complexes by a Clq/protein A-ELISA during the pre- 91. de Mari K, Maynard L, Sanquer A, et al. Therapeutic effects of
neoplastic stages of feline leukemia virus infection. Vet Immunol recombinant feline interferon-omega on feline leukemia virus
Immunopathol. 1984;7:227-238. (FeLV)-infected and FeLV/feline immunodeficiency virus (FIV)-
70. Day NK, O’Reilly-Felice C, Hardy Jr WD, et  al. Circulating coinfected symptomatic cats. J Vet Intern Med. 2004;18:477-482.
immune complexes associated with naturally occurring lympho- 92. Hartmann K, Donath A, Beer B, et  al. Use of two virustatics
sarcoma in pet cats. J Immunol. 1980;125:2363-2366. (AZT, PMEA) in the treatment of FIV and of FeLV seroposi-
71. Snyder Jr HW, Jones FR, Day NK, et  al. Isolation and charac- tive cats with clinical symptoms. Vet Immunol Immunopathol.
terization of circulating feline leukemia virus-immune complexes 1992;35:167-175.
from plasma of persistently infected pet cats removed by ex vivo 93. Nelson P, Sellon R, Novotney C, et al. Therapeutic effects of dieth-
immunosorption. J Immunol. 1982;128:2726-2730. ylcarbamazineand3′-azido-3′-deoxythymidineonfelineleukemiavirus
72. Fails AD, Mitchell TW, Rojko JL, et  al. An oligopeptide of the lymphomaformation.VetImmunolImmunopathol.1995;46:181-194.
feline leukemia virus envelope glycoprotein is associated with 94. Cattori V, Weibel B, Lutz H. Inhibition of feline leukemia virus
morphological changes and calcium dysregulation in neuronal replication by the integrase inhibitor raltegravir. Vet Microbiol.
growth cones. J Neurovirol. 1997;3:179-191. 2011;152:165-168.
238 SECTION 1  Viral Diseases

95. Addie DD, Dennis JM, Toth S, et al. Long-term impact on a closed 99. Jirjis F, Davis T, Lane J, et  al. Protection against feline leuke-
household of pet cats of natural infection with feline coronavirus, mia virus challenge for at least 2 years after vaccination with
feline leukaemia virus and feline immunodeficiency virus. Vet Rec. an inactivated feline leukemia virus vaccine. Vet Ther. 2010;11:
2000;146:419-424. E1-E6.
96. Vail DM, Moore AS, Ogilvie GK, et  al. Feline lymphoma (145 100. Richards JR, Elston TH, Ford RB, et  al. The 2006 American
cases): proliferation indices, cluster of differentiation 3 immuno- Association of Feline Practitioners Feline Vaccine Advisory Panel
reactivity, and their association with prognosis in 90 cats. J Vet report. J Am Vet Med Assoc. 2006;229:1405-1441.
Intern Med. 1998;12:349-354. 101. Lutz H, Addie D, Belak S, et  al. Feline leukaemia. ABCD guide-
97. Sykes JE, et al, unpublished observations. 2012. lines on prevention and management. J Feline Med Surg. 2009;11:
98. Hofmann-Lehmann R, Cattori V, Tandon R, et  al. Vaccination 565-574.
against the feline leukaemia virus: outcome and response categories
and long-term follow-up. Vaccine. 2007;25:5531-5539.
CHAPTER 23

Feline Respiratory Viral Infections


Jane E. Sykes

lifetime. FHV-1 is an alphaherpesvirus that is closely related


Overview of Feline Viral Respiratory Disease to canine herpesvirus-1, and to a lesser extent, herpes simplex
First Described in Cats: Feline herpesvirus-1 (FHV-1), 1958 virus. There is only a single serotype of FHV-1. Isolates are also
(United States)1; feline calicivirus (FCV), 1957 (New Zea- genetically similar, yet some variation in strain virulence exists.6
land)2; H5N1 influenza virus, 2006 (Thailand)3; pandemic Using culture, FHV-1 has been detected in 0% to 39% of
H1N1 influenza virus, 2010 (United States).4 cats with URTD, although in some catteries and shelters with
endemic FHV-1 infection, the prevalence may be much higher.
Causes: Feline herpesvirus-1, feline calicivirus, avian and
When sensitive PCR assays are used to detect FHV-1, prevalences
human origin influenza A viruses
of infection that approach 100% have been detected in some
Geographic Distribution: Worldwide groups of cats with acute URTD.7 The prevalence of shedding by
Mode of Transmission: Close contact, fomite spread, to a apparently healthy cats has ranged from 0% to 10% and most
lesser extent aerosols often has been lower than 2%.5,8-14 Virtually all infected cats
develop latent infection, which primarily occurs in the trigeminal
Major Clinical Signs: Serous to mucopurulent nasal and ocu-
ganglia. FHV-1 DNA can also be detected in other tissues of the
lar discharge, sneezing, stertor, conjunctivitis, lingual or
head, such as the cornea and nasal cavity, but whether this is
facial ulceration, fever, inappetence, tachypnea, cough,
a true state of latency or just chronic persistent infection is not
rarely death. Keratitis occurs with FHV-1 infection.
clear. Reactivation of shedding, with or without concurrent clin-
Differential Diagnoses: Infections with bacteria such as Borde- ical signs of URTD, occurs in less than half of latently infected
tella bronchiseptica, Streptococcus spp., or Chlamydia felis; cats 4 to 12 days after stress.15 Examples of stressors include
other causes of rhinitis such as cryptococcosis, aspergillo- transportation (such as to a veterinary clinic, boarding or breed-
sis, neoplasia, foreign bodies, chronic idiopathic feline rhi- ing facility, shelter, or cat show), lactation, exposure to new cats,
nosinusitis, nasopharyngeal stenosis, or nasopharyngeal concurrent illness, or treatment with immunosuppressive drugs
polyps; feline chronic airway disease. such as glucocorticoids. The duration of shedding after reactiva-
Human Health Significance: Influenza virus infections have tion ranges from 1 to 13 days (mean, 7 days).16,17 Shedding that
been transmitted from humans to cats, and the potential coincides with lactation results in infection of susceptible kittens.
for zoonotic transmission exists. FHV-1 and FCV do not FHV-1 survives a maximum of 18 hours at room tempera-
infect humans. ture and is readily inactivated by drying and most disinfectants.
Because of this, transmission occurs primarily through close
contact, although fomites remain a very important mode of
transmission in crowded environments. Aerosol transmission is
Etiology and Epidemiology of lesser importance.15,18 Aerosols generated by sneezing cats
typically travel no further than a distance of 4 to 5 feet.
Infectious feline upper respiratory tract disease (URTD) is a
widespread and important cause of morbidity and mortality Feline Calicivirus
where large numbers of cats are housed together, especially Feline calicivirus (FCV) is a non-enveloped, single-stranded
in overcrowded or stressful conditions.5 Multiple pathogens RNA virus with a spherical capsid that is studded with cup-
are involved and co-infections are common (Table 23-1). The shaped depressions (calici = “cup”) (Figure 23-1). Like FHV-1,
most prevalent viral causes of URTD are feline herpesvirus-1 FCV commonly causes feline URTD, accounting for 10% to
­(FHV-1) and feline calicivirus (FCV). Influenza viruses also over 50% of cases. The highest prevalences occur in large multi-
cause respiratory disease in cats but are relatively rarely iden- cat environments. Natural infection of dogs by FCV-like strains
tified. Bacterial causes of respiratory disease in cats include has been reported.19 Like other RNA viruses, the genome of
Bordetella ­bronchiseptica, Chlamydia felis, and Mycoplasma FCV continually undergoes rapid mutation, which increases
species. Streptococcus canis and Streptococcus equi subspecies the diversity of strains over time. Although considerable anti-
zooepidemicus can also play a role in shelter situations and cat- genic diversity exists among FCV isolates, the degree of anti-
teries. More information on the bacterial causes of feline URTD genic cross-reactivity between them is sufficient for them to be
can be found in Chapters 33, 34, 38, and 40. classified as a single serotype. Nucleotide sequence analysis also
suggests that isolates worldwide are a single, but highly diverse,
Feline Herpesvirus-1 group. No correlations have been found between the genetic
FHV-1 is a large, enveloped DNA virus that has a worldwide composition of FCV strains, different clinical manifestations of
distribution. Most cats are exposed to FHV-1 during their disease, or geographical location.

239
240 SECTION 1  Viral Diseases

TABLE 23-1
Viral Causes of Transmissible Respiratory Disease in Cats
Organism Incubation Period (days)* Shedding Period* Environmental Survival
Feline herpesvirus-1 2 to 6 1 to 2 weeks, with intermittent Less than a day
­reactivation of shedding with stress
Feline calicivirus 2 to 6 Most <30 days, may be lifelong Up to 1 month
Influenza viruses Few days <2 weeks Hours

*Incubation and shedding periods are approximate and may differ when co-infections or immune suppression operate.

often affected, whereas kittens tend to show less severe signs.


Although infections typically spread rapidly in outbreaks,
including through fomites to pet cats of hospital staff, spread
of disease has been limited to affected clinics or shelters, with
no further spread within the community, and outbreaks resolve
within approximately 2 months once appropriate control mea-
sures are instituted.

Influenza Viruses
Pandemic H1N1 influenza viruses originated from pigs and
have caused widespread illness in humans. Natural infection
with these strains has been reported in cats from the United
FIGURE 23-1  Schematic of the structure of the capsid of feline calicivirus. States and Europe. Affected cats had signs of fever and respi-
ratory and gastrointestinal tract disease and, in some cases,
Infected cats can develop a persistent oropharyngeal infec- death.4,33-35 Transmission from sick humans who were in con-
tion (>1 month in duration) in the absence of obvious clinical tact with the cats was suspected. Cat-to-cat transmission may
signs. This is termed the carrier state for FCV, and may result also have occurred.34
from immune evasion through antigenic variation of the capsid Highly pathogenic avian origin H5N1 influenza virus infec-
protein.20 FCV is shed continuously from the oropharynx, and tion, which emerged as a cause of human illness in Hong
the magnitude of shedding varies with time and between indi- Kong in 1997 and subsequently spread worldwide,36 has been
vidual cats.21 Carrier cats serve as a source of infection for other detected in sick and apparently healthy domestic cats and wild
cats. In many cats, shedding terminates weeks to months after felids from southeast Asia3,37,38 and central Europe.39,40 Nev-
infection, but in a few cats, shedding is lifelong. A single cat ertheless, no evidence of infection was found in more than 170
can be simultaneously infected with multiple variants of FCV, cats where infected birds had been identified in Germany and
each derived from the original infecting strain as a result of Austria.41 Infection of cats follows direct or indirect contact
genetic mutation, drift, and selection pressures.20 Because of the with infected birds, especially consumption of raw poultry;
chronic carrier state, the prevalence of FCV infection in healthy ­cat-to-cat ­transmission may also occur.37
cats is high and ranged from 8% of household cats to 24% of Canine H3N2 virus infection was associated with bron-
show cats.12 chopneumonia in shelter-housed cats in China and Korea.42
Environmental persistence of FCV is considerably more pro- Experimentally, cats can also be infected with other influenza
longed than that of FHV-1, and FCV resists routine disinfection viruses, including human H2N2 and H3N2, and avian origin
with quaternary ammonium compounds. Susceptibility to disin- H7N7 and H7N3 viruses.43,44 Widespread evidence of exposure
fectants may vary between FCV strains.22 Survival in the envi- to pandemic H1N1, seasonal H1N1, and seasonal H3N2 was
ronment has been demonstrated for as long as 28 days,23 and found in 400 cats from Ohio in the United States.45 Seropositive
related caliciviruses persist in a dried state for several months.24 cats were 7.4 times more likely to have had respiratory illness
As a result, fomites are a very important means of transmission. than seronegative cats, and twice as likely to have had nonrespi-
FCV is also transmitted through direct contact with respiratory ratory illness than seronegative cats. More information on influ-
secretions and through aerosols. Fleas may spread FCV through enza viruses can be found in Chapter 17.
their feces or when cats ingest fleas while grooming.25
Highly virulent FCV strains have been isolated from out- Clinical Features
breaks of severe systemic febrile illness in cats in the United
States and Europe known as virulent systemic disease (VSD),26-31 Signs and Their Pathogenesis
which was first described in California in 1998. Shelter cats After direct or indirect contact with virus in respiratory secre-
that were hospitalized in veterinary clinics have been a source tions from the conjunctiva, nasal cavity and oropharynx, FCV
of infection in many outbreaks, and for each outbreak, the FCV and FHV-1 replicate within lymphoid and epithelial cells in the
strain involved has differed. VSD has also been described in upper respiratory tract and cause cytolysis. FHV-1 also repli-
one cat from a multiple-cat household, that is, in the absence cates in corneal epithelial cells.46 Although FHV-1 prefers to
of an outbreak.32 Otherwise healthy, adult, vaccinated cats are replicate in the cooler tissues of the upper respiratory tract,
CHAPTER 23  Feline Respiratory Viral Infections 241

systemic infection with viremia occurs in some cats infected with


FHV-1 and may be more likely to occur in neonates and debili-
tated cats. FCV and influenza viruses also replicate systemically.
FCV is shed in the urine and feces of infected cats, in addition to
respiratory secretions. Influenza viruses are shed in both respi-
ratory secretions and the feces. Avian origin H5N1 influenza
viruses replicate initially in the lower respiratory tract (type
II pneumocytes and alveolar macrophages) or ­gastrointestinal
tract (for example, after ingestion of an infected bird), which is
followed by severe systemic infection with necrosis and inflam-
mation in multiple organs.38 In contrast, severe systemic infec-
tion is not a feature of H1N1 influenza virus infections in cats.47
Clinical signs of viral URTD occur after an incubation
period of 2 to 6 days, although longer incubation periods are
also possible, and incubation periods as short as 1 to 2 days
can occur in cats infected with influenza viruses.47 Viral shed-
A
ding occurs as early as 24 hours after infection, and often before
the onset of clinical signs. Clinical signs range considerably in
severity from no signs or mild serous ocular discharge to pneu-
monia and death. The most severe signs tend to occur in very
young or elderly debilitated cats. Concurrent immunosuppres-
sive illness or infection with other respiratory pathogens and
opportunistic bacteria also profoundly influences disease sever-
ity. Clinical signs include conjunctivitis, serous or mucopurulent
ocular and nasal discharge and sneezing, and, less commonly,
­cough or tachypnea (Figure 23-2, A). Mucopurulent discharges
result from secondary bacterial infections with opportunistic
pathogens such as Streptococcus spp., Staphylococcus spp., Pas-
teurella multocida, and Escherichia coli. Ocular and nasal dis-
charges may become crusted, and kittens’ eyes may not open as
a result of the sticky exudates; when severe this can be followed
by extensive corneal damage and rupture of the globe. Lethargy, B
inappetence, hypersalivation, and fever may be present in acute FIGURE 23-2  Chemosis, mucopurulent ocular and nasal discharge (A) and lingual
infections. FCV and FHV-1 infections may also lead to pharyn- ulceration (B) in a 6-month-old intact male domestic medium-hair cat with chronic
gitis and laryngitis, which can be accompanied by clinical signs nasal discharge and conjunctivitis. A conjunctival swab specimen tested positive with
of gagging or obstructive respiratory patterns. Ulcerative glos- a PCR assay for FCV RNA and negative for FHV-1, Chlamydia felis, and Mycoplasma
sitis and ulceration of the nasal planum, conjunctiva, and skin spp. DNA. (Courtesy of the University of California, Davis Veterinary Ophthalmology
is more common and severe with FCV infection but can also be Service.)
associated with FHV-1 infection (see Figure 23-2, B). Clinical
features unique to each infection are outlined next.
also cause a severe ulcerative and eosinophilic facial dermatitis
Feline Herpesvirus-1 ­(Figure 23-3). Lesions have also been described elsewhere on the
In neonatal infections caused by FHV-1, damage to upper respi- body in the absence of facial lesions.49
ratory epithelium may lead to osteolysis of the nasal turbinates
and persistent or recurrent sinusitis and rhinitis. Rarely, neuro- Feline Calicivirus
logic signs and reproductive complications such as abortion and FCV infection has been most strongly associated with erosive or
fetal resorption have been observed in infected cats, although ulcerative lesions, which can occur on the nasal planum, tongue,
it is unclear what role FHV-1 itself plays in the pathogenesis lips, and occasionally the conjunctiva and heal over a period of
of these clinical manifestations. FHV-1 is an important cause 2 to 3 weeks.50 Persistent infection with FCV has also been
of corneal disease in cats, and has been implicated as a cause of linked to chronic ulceroproliferative and lymphoplasmacytic
acute and chronic ulcerative (epithelial) keratitis, stromal kera- stomatitis, which involves the mucosa lateral to the palatoglos-
titis, eosinophilic keratitis, corneal sequestra, and uveitis. Stro- sal arches (caudal stomatitis), the alveolar mucosa in the premo-
mal keratitis is thought to be an immunopathologic response lar and molar area, and sometimes the buccal mucosa (alveolar/
to persistent viral antigens. The presence of dendritic corneal buccal mucositis) (Figure 23-4).51-53 Some persistently infected
ulcers is thought to be pathognomonic for FHV-1 infection. cats have isolated hyperemia of the buccal mucosa along the
However, the role that FHV-1 plays as a cause of eosinophilic length of the dental arcade in the absence of significant peri-
keratitis, corneal sequestra, and uveitis requires further study, odontal disease. There is no age predisposition for this condi-
because a clear association between these abnormalities and tion.53 Pyrexia and transient lameness due to synovitis has been
the detection of FHV-1 within corneal tissues has not always described days to weeks after clinical signs of acute FCV infec-
been present.48 Consequences of ocular disease due to FHV-1 tion and after vaccination with certain FCV vaccines. FCV has
include symblepharon (adhesion of an ulcerated conjunctiva to been investigated as a possible cause of feline lower urinary tract
itself or the cornea) and keratoconjunctivitis sicca. FHV-1 can disease (feline interstitial cystitis) and enteritis in cats. Because
242 SECTION 1  Viral Diseases

FIGURE 23-4  Severe caudal stomatitis in a retrovirus-negative, 8-year-old female


spayed domestic shorthair cat that was evaluated for ptyalism. The cat lived with 12 other
cats. Histopathology revealed severe plasmacytic and neutrophilic inflammation with
multifocal epithelial ulceration and hyperplasia.

oral and footpad ulceration, and nasal and/or ocular discharge.


VSD strains infect not only epithelial cells of the upper respira-
tory tract and oral cavity, but a variety of other cell types, such
as endothelial cells, hepatocytes, pneumocytes, and pancreatic
acinar cells.54 FCV uses feline junctional adhesion molecule
A (JAM-A) as a receptor, a member of the immunoglobulin
B superfamily. The lesions that develop are thought to result from
disruption of intercellular tight junctions and vasculitis. Distinc-
tive clinical signs of VSD include cutaneous edema, alopecia,
crusting, and ulceration. Edema occurs most commonly on
the head and limbs but may become generalized. Crusting and
ulceration are most prominent on the nose, lips, pinnae, peri-
ocular regions, and distal limbs. Severe respiratory distress due
to pulmonary edema or pleural effusion, or icterus as a result of
hepatic necrosis or pancreatitis, develop in some cats and have
been associated with a poor prognosis. Involvement of the gas-
trointestinal tract, liver, and pancreas may also result in vomit-
ing and/or diarrhea. Cats also develop a coagulopathy, which
can be manifested by petechial and ecchymotic hemorrhages
and, rarely, epistaxis and hematochezia. In peracute infections,
cats die as a result of cardiovascular arrest with few preceding
C
signs apart from fever.
FIGURE 23-3  Ulcerative and eosinophilic facial dermatitis secondary to FHV-1
infection. A, 3-year-old female spayed domestic shorthair with a 2-month history of a Influenza Virus Infections
crusted lesion on the right dorsal muzzle. B, Fourteen-year-old female spayed domes- In addition to fever, anorexia, lethargy, conjunctivitis, nasal and
tic shorthair with blepharokeratoconjunctivitis and severe ulcerative facial dermatitis.  ocular discharges, and tachypnea as a result of viral pneumonia,
C, Same cat as in B after 4 months of treatment with famciclovir. Fluorescein stain is pres- highly pathogenic H5N1 avian origin influenza virus infections
ent. Biopsy in both cats showed severe, diffuse, necrotizing, and eosinophilic dermatitis
have been associated with neurologic signs such as seizures and
with a few intranuclear inclusions bodies. A PCR assay for FHV-1 DNA on a biopsy from the
cat in B was positive. (Courtesy of the University of California, Davis Veterinary Dermatol- ataxia, which result from nonsuppurative meningoencephalitis
ogy and ­Ophthalmology Services.) and vasculitis.36 Diarrhea and vomiting have not been observed
in cats infected with H5N1 viruses. Vomiting has been reported
in cats infected with pandemic H1N1 influenza viruses, which
FCV can be shed in the urine and feces of apparently healthy may have a predilection for the gastrointestinal tract.33,34
cats, the significance of the virus in the pathogenesis of these
conditions remains unclear. Physical Examination Findings
Cats with VSD show severe signs of caliciviral URTD, Physical examination findings in cats with acute viral URTD
including anorexia, fever (often >105°F [40.6°C]), weight loss, vary from mild serous ocular discharge and conjunctivitis
CHAPTER 23  Feline Respiratory Viral Infections 243

through to fever, severe mucopurulent ocular and nasal dis-


charges, chemosis, dehydration, thin body condition, stertorous
or stridorous respiration, tachypnea, increased breath sounds
on thoracic auscultation, hypersalivation, and ulceration of the
nasal planum, tongue, and lips. Because ulcerative lesions can
occur at the base of the tongue near the larynx, examination of
the entire tongue should be performed in cats that are febrile
and inappetent, which may require sedation. Cats with VSD
can have edema of the face and lips, icterus, cutaneous ulcer-
ation, evidence of petechial hemorrhages, and abdominal pain.
Ulcerative keratitis with dendritic or geographical corneal ulcer-
ation may be seen in cats with acute FHV-1 infection; chronic
FHV-1 infection may be manifested as stromal keratitis with
neovascularization, pigmentation and fibrosis of the cornea,
symblepharon, and conjunctivalization of the cornea (Figure
23-5). Eosinophilic keratitis manifests as superficial, pink to
A
white vascularized proliferative lesions on the cornea or con-
junctiva; abundant eosinophils are present in smears of corneal
scrapings. Herpetic facial dermatitis is manifested as cutane-
ous ulceration, erythema, exudation, and adherent crusts, most
commonly around the nose and eyes but occasionally on the
trunk and limbs.55 Cats with persistent FCV infection may have
ulceroproliferative caudal stomatitis or buccal/alveolar mucosi-
tis, and exhibit pain on examination of the oral cavity.

Diagnosis
It is not always possible to identify the cause of transmissible
respiratory disease in cats based on clinical signs alone, because
each pathogen produces a similar spectrum of signs. The pres-
ence of corneal ulceration raises suspicion for FHV-1 infection, B
and severe lingual ulceration or facial edema and crusting raises
suspicion for FCV infection, but mixed infections occur and
complicate diagnosis. A history of exposure to other cats pro-
vides support for a diagnosis of viral respiratory disease, but
because FHV-1 infections can recrudesce with stress, potential
or known exposure to other cats is not essential to implicate
respiratory viruses as a cause of disease. Previous immunization
for feline respiratory viruses does not rule out the possibility of
viral URTD. All cats with signs of URTD should have known
retrovirus infection status, because viral URTD is common in
retrovirus-infected cats. When signs such as chronic nasal dis-
charge are present, other etiologies should also be considered,
such as fungal infections, neoplasia, and foreign bodies. Diag-
nosis of eosinophilic keratitis and ulcerative facial dermatitis
may require corneal scrapings or skin biopsy, respectively. C
FIGURE 23-5  A, Ulcerative and stromal keratitis in a 2-year-old male neutered rex
Laboratory Abnormalities cat. FHV-1 infection was suspected. Treatment with famciclovir (375 mg PO q8h), l-lysine
There are no specific CBC, biochemistry profile, or urinalysis (500 mg PO q12h), and cidofovir (0.5%; one drop OD q12h) was associated with clinical
abnormalities that aid in a diagnosis of feline viral respiratory improvement over a 1 month period (B and C). The cat had a history of a corneal seques-
disease. The CBC can be normal or show a mild to moderate trum and subsequently developed eosinophilic keratitis. (Courtesy of the University of
neutrophilia, sometimes with band neutrophils or neutrophil California, Davis Veterinary Ophthalmology Service.)
toxicity. Lymphopenia may be present in severely affected
cats. Serum biochemistry findings in cats with viral respiratory Tracheobronchial lavage specimens from cats with pneu-
disease are usually unremarkable unless disease is severe or monia may show a suppurative or mixed exudate, sometimes
chronic. In cats with VSD, hematologic abnormalities include with evidence of secondary bacterial infection. Aerobic bacterial
mild to severe anemia, thrombocytopenia, neutrophilia, and culture and susceptibility testing and culture for Mycoplasma
lymphopenia. Cats with VSD may also have hypoalbuminemia, spp. are indicated on wash specimens. Organisms such as Pas-
hyperbilirubinemia, mildly increased serum activities of ALT teurella spp., Staphylococcus pseudintermedius, Streptococcus
and AST, and increased serum CK activity. Serum CK activities spp., Escherichia coli, Klebsiella pneumoniae, and some Myco-
up to 11,000 U/L can occur.26 Cats with chronic stomatitis may plasma spp. infect the airways as opportunists. Bacterial culture
have hyperglobulinemia due to a polyclonal gammopathy. of nasal swabs is generally not recommended because it often
244 SECTION 1  Viral Diseases

TABLE 23-2
Diagnostic Assays Available for Respiratory Virus Infections in Cats
Assay Specimen Type Target Performance
Virus isolation Conjunctival, nasal, and caudal Virus Negative results can occur if specimens contain
pharyngeal swabs, transtra- no or low numbers of virus particles. May take
cheal and bronchoalveolar several days and requires specialized techniques
wash specimens, airway and and expertise. May be the most sensitive assay for
lung specimens collected at detection of FCV infections.
necropsy
PCR See Virus isolation Viral nucleic acid Sensitivity and specificity may vary depending on as-
say design. Sensitivity may be low because of brief
shedding for some viruses or low-level shedding in
chronic infections. Assays may not detect all virus
strains. Testing specimens from multiple different
anatomic sites or combining PCR with other diag-
nostic tests can increase sensitivity. Attenuated live
vaccine virus may be detected after vaccination.
Because of subclinical shedding, the significance
of a positive result may be difficult to interpret.
False-negative results may occur as a result of
degradation of viral nucleic acid during specimen
transport.
Serology Serum Antibodies against Interpretation complicated by previous vaccination
respiratory viral and exposure. Acute and convalescent serology
antigens may be useful in outbreak situations that involve a
novel strain of FCV or influenza A virus.
Histopathology Necropsy or biopsy ­specimens FHV-1 inclusions; Detection of FHV-1 inclusions has low sensitivity,
immunostaining and immunostaining and/or PCR is required to
definitively identify the infecting virus.

FCV, Feline calicivirus; FHV-1, feline herpesvirus-1.

leads to growth of normal flora, which has no clinical relevance. severe disease, necropsies can provide valuable information and
However, resistant Pseudomonas aeruginosa infections can should be performed by a veterinary pathologist as soon as pos-
develop in some cats with chronic URTD that have been treated sible after death or euthanasia. Tissues should be submitted for
repeatedly with antimicrobial drugs. histopathology (in formalin), bacterial and virus isolation (fresh
tissue), and PCR for respiratory viruses and bacteria (fresh or
Diagnostic Imaging frozen tissue; see Chapter 5). There is currently no way to dis-
In uncomplicated feline URTD viral infections, plain thoracic tinguish FCV strains that cause VSD from other FCV strains.
radiographs may be unremarkable or show a mild diffuse Assays available to detect feline respiratory viruses include
interstitial to bronchointerstitial pattern. Alveolar patterns or virus isolation, direct immunofluorescence, and PCR assays
lung lobe consolidation can occur with secondary bacterial (Table 23-2). Point-of-care ELISA assays developed for use in
pneumonia. humans have been evaluated for detection of H5N1 influenza
virus antigen in cats56 but have low sensitivity and will not be
Microbiologic Tests discussed further. Because influenza viruses and FCV are RNA
Because many cats experience self-limiting disease, attempts to viruses that exhibit considerable sequence diversity, virus iso-
obtain an etiologic diagnosis should be made when disease per- lation may offer the greatest sensitivity for detection of these
sists for longer than 7 to 10 days or is complicated by pneumo- viruses and enables typing for influenza viruses. In contrast,
nia, with lethargy and inappetence. When outbreaks occur in FHV-1 loses infectivity more rapidly than FCV and has little
shelters or the pattern of endemic respiratory disease changes, sequence diversity, so PCR is more useful. The use of both
attempts to make an etiologic diagnosis are also indicated and PCR and culture together offers the greatest sensitivity when
strongly encouraged. Because FCV and FHV-1 can be detected it is important to obtain a diagnosis. Serology may be of use
in apparently healthy cats, it may be difficult to know the sig- for investigation of outbreaks that involve novel FCV strains or
nificance of a positive test result in a single cat with signs of influenza virus types.
respiratory disease. In an outbreak situation, collection of mul-
tiple specimen types from several cats with and without clini- Virus Isolation
cal signs can facilitate diagnosis and allow interpretation of the FCV, FHV-1, and influenza viruses are readily isolated in cell
significance of test results. In shelter situations or outbreaks of culture. Virus isolation for diagnosis of feline respiratory viral
CHAPTER 23  Feline Respiratory Viral Infections 245

infections is offered to veterinarians by some commercial vet- severe histologic lesions.62 In the future, quantitation of viral
erinary diagnostic laboratories that specialize in virology. The nucleic acid within swabs may assist interpretation of the clinical
laboratory should be notified if influenza virus infection is a pos- significance of positive PCR results. PCR can also detect attenu-
sibility. Suitable specimens for isolation of respiratory viruses ated live vaccine virus that is shed after routine vaccination. How-
include conjunctival, nasal, and oropharyngeal swabs, transtra- ever, this appeared to be an uncommon phenomenon in one study
cheal or bronchoalveolar lavage specimens, or upper airway tis- that used PCR to detect FHV-1 and FCV in 12 cats over a 3-week
sue or lung obtained at necropsy. Influenza viruses may also be period after vaccination with intranasal or parenteral attenuated
isolated from feces or rectal swabs. If swabs are used for speci- live FHV-1 and FCV vaccines. 14 The cats in this study had been
men collection, polyester-tipped swabs and specific virus trans- previously exposed to FHV-1, and two had been vaccinated with
port media should be used, which are available from commercial FHV-1 and FCV vaccines, which may have reduced shedding
testing laboratories. Cotton swabs should be avoided, because after vaccination. Additional studies are required to determine
influenza viruses adhere to the cotton, which may lead to the prevalence of positive assay results after vaccination and their
reduced sensitivity.57 When conjunctival swabs are collected, the relationship to vaccine virus. In outbreak situations, false-positive
use of topical anesthetics should also be avoided, because they PCR results have the potential to occur if swabs become contami-
have the potential to reduce the sensitivity of virus isolation.58 nated with virus from the environment or the hands of personnel.
Isolation may take several days. Sensitivity can be very low in Clean examination gloves should be worn for each cat, and the
cats with chronic manifestations of infection (for example, more swab should touch only the anatomic site to be tested.
than 1 week after the onset of clinical signs), because shedding
of respiratory viruses can be transient. In cats with mixed FCV Serologic Diagnosis
and FHV-1 infections, FCV may obscure the presence of FHV-1, Serologic assays (such as ELISA or serum neutralization assays)
because it produces cytopathic effects more rapidly. that detect antibodies to FHV-1 and/or FCV are offered on a
commercial basis to veterinarians. Unfortunately, serology is
Fluorescent Antibody Testing not useful for diagnosis, because of vaccine titer interference
Fluorescent antibody that specifically detects FHV-1 or FCV and the high prevalence of subclinical exposure to these viruses
can be applied to conjunctival smears or impression smears in the cat population. Titers to FCV may vary depending on the
from lung tissue collected at necropsy. The effect of vaccination degree of homology between the infecting virus and the FCV
with attenuated live vaccines on fluorescent antibody test results strain used in the assay. Nevertheless, serology has been useful
has not been reported. The sensitivity of fluorescent antibody to investigate some outbreaks of VSD. Serology has also been
testing is lower than that of PCR and virus isolation,59 and inex- used to predict protection from infection, although cats with
perienced laboratory personnel may misinterpret nonspecific negative titers can still have some degree of immunity, and cats
fluorescence as a positive result. with positive titers may develop illness after challenge.
Serologic assays for influenza virus exposure are based
Molecular Diagnosis Using the Polymerase Chain Reaction on serum neutralization or hemagglutination-inhibition (see
Panels of real-time PCR assays that detect feline respiratory Chapter 2) and are specific for the influenza virus strain of inter-
pathogens are offered by some commercial veterinary diagnos- est (i.e., H5N1, H1N1).
tic laboratories and are rapid and relatively inexpensive. These
can include assays for FHV-1 and FCV, as well as bacteria such Pathologic Findings
as Mycoplasma spp., B. bronchiseptica, and C. felis. RT-PCR Gross lesions in cats with viral URTD include cutaneous and lin-
assays for the detection of influenza viruses are also available. gual ulcerations, evidence of keratitis in cats infected with FHV-
The sensitivity and specificity of PCR assays can vary consider- 1, submandibular or retropharyngeal lymph node enlargement,
ably between laboratories. Swabs of the nasal cavity, conjuncti- mucosal hyperemia, and mucopurulent exudate within the nasal
val sac, or caudal pharynx; tracheobronchial lavage specimens; cavity and trachea. In cats with pneumonia, the lungs may be
skin biopsies; or upper respiratory tract and lung tissue col- edematous with diffuse or multifocal consolidated areas. Histo-
lected at necropsy are suitable specimens for testing. Topical pathology may show fibrinosuppurative and necrotizing stomati-
anesthetics and fluorescein can reduce the sensitivity of PCR tis, rhinitis, tracheitis, and/or alveolitis. When FHV-1 is present,
assays for human herpesviral infections,60 and so they should intranuclear viral inclusions may be found within epithelial cells
be avoided, although one study showed no inhibitory effect of the upper respiratory tract (Figure 23-6). Osteolytic changes
of topical fluorescein or proxymetacaine in cats on FHV-1 or within the nasal turbinates may also be identified in cats infected
C. felis PCR results.61 False-negative PCR results can occur in with FHV-1. Virus can be detected in epithelial cells using immu-
cats with chronic infections that shed at low levels. Because FCV nohistochemistry or fluorescent antibody staining. Skin biopsies
and influenza viruses are RNA viruses, false negatives may also from cats with ulcerative herpesviral dermatitis show epidermal
result from degradation of viral RNA during specimen trans- ulceration and necrosis that extends into the superficial dermis,
port or as a result of strain variation. and infiltration with degenerated neutrophils, eosinophils, and
Positive PCR assay results for FCV and FHV-1 should be fewer numbers of histiocytes, plasma cells, and lymphocytes. In
interpreted with caution, because apparently healthy cats can some skin biopsies, intranuclear viral inclusions are identified.55
shed these viruses and, although controversial, FHV-1 DNA Necropsy findings in cats with VSD are variable. Frequently
detected in corneal, conjunctival, or nasal biopsy tissue may rep- reported findings include individual hepatocellular necrosis and
resent latent virus. In one shelter, a correlation between the quan- dissociation with minimal inflammation, acute interstitial pneu-
tity of FHV-1 present in pooled oropharyngeal and conjunctival monia, and free pleural and abdominal fluid.26-28,31 Acute nec-
swabs as determined using quantitative real-time PCR and the rotizing vasculitis of subcutaneous and oral submucosal vessels
severity of histologic lesions within the nasal cavity was detected, has been described.32 Intestinal crypt necrosis and pancreatitis
with cycle threshold values less than 29 correlating with the most have been reported in experimentally infected cats.
246 SECTION 1  Viral Diseases

Herpetic Keratoconjunctivitis and Facial


Dermatitis
The use of antiviral drugs should be considered for cats with
severe and recurrent or persistent manifestations of FHV-1
infection such as keratitis, severe conjunctivitis, and ulcerative
facial dermatitis. Antiviral drugs are discussed in more depth
in Chapter 7 (see Table 7-2 for dosages). Referral to a veteri-
nary specialist for assessment of an underlying cause should
be considered for cats with signs of chronic URTD. Acyclovir
and its prodrug, valacyclovir, cause unacceptable toxicity when
administered systemically to cats, and so their use is not recom-
mended. In contrast, oral famciclovir is well tolerated by adult
cats and kittens and results in significant clinical improvement
and decreased viral shedding in cats with acute and chronic her-
pesviral disease (see Figure 23-3, B and C).63,64 The prodrug of
A 1 mm famciclovir, penciclovir, also has potential to be a useful drug
for treatment of feline herpesviral infections. Because FHV-1
infection can deplete conjunctival goblet cells,64a frequent appli-
cation of a topical mucinomimetic that contains hyaluronic acid
is recommended in conjunction with famciclovir.
Topical ophthalmic preparations that contain idoxuridine,
trifluridine, vidarabine, or acyclovir can be used to treat herpes-
viral keratitis, although the true efficacy of these drugs has not
been well studied. Frequent topical application is required (5 to
6 times daily), and prolonged use can cause corneal irritation or
ulceration. Idoxuridine and vidarabine are better tolerated by
cats than trifluridine; vidarabine is effective against idoxuridine-
resistant strains. Cidofovir and ganciclovir show greater prom-
ise as topical treatments and are highly active against FHV-1
in  vitro. Topical cidofovir decreases disease severity and viral
shedding in experimentally infected cats,65 and has the advan-
B tage of requiring only twice daily administration.
Human recombinant IFN-α and recombinant feline IFN-ω
FIGURE 23-6  A, Cross-section of the nares of a shelter cat with rhinitis due to infec- inhibit FHV-1 replication in vitro and have been administered
tion by FHV-1. Ulceration is most severe on the right side. Hematoxylin and eosin stain. parenterally, topically (for keratitis), and orally to cats with
B, Higher magnification of the nasal epithelium that shows rhinitis and the presence of
FHV-1 infection. Obvious clinical responses to treatment have
intraepithelial, intranuclear inclusion bodies. (From Burns RE, Wagner DC, Leutenegger
CM, et al. Histologic and molecular correlation in shelter cats with acute upper respiratory not been uniformly observed, but controlled clinical trials that
infection. J Clin Microbiol 2011;49(7):2454-2460. Images courtesy Dr. Patricia Pesavento, involve large numbers of cats are lacking. Improvement was
University of California, Davis Veterinary Anatomic Pathology Service.) noted when IFN-α was given subcutaneously 2 days before
inoculation of cats with FHV-1,66 but the effect of parenteral
IFN-α after the onset of clinical signs requires further investiga-
In addition to acute respiratory lesions, cats infected with tion. Parenteral administration may be followed by the develop-
H5N1 influenza viruses can have widespread necrosis and ment of neutralizing antibodies beyond 3 weeks of treatment.
inflammation in many organs. Lysine has shown efficacy for treatment of herpesviral con-
junctivitis in cats when administered as tablets67 and can reduce
Treatment and Prognosis reactivated shedding by latently infected cats.68 However, when
administered to shelter cats, there was no reduction in URTD,
Acute Upper Respiratory Disease possibly because of the stress of tablet administration.69 Dietary
Supportive care is the mainstay of treatment for acute feline formulation with lysine has also not been effective for manage-
URTD and includes subcutaneous fluid therapy, antimicrobial ment of URTD in shelters and in fact may contribute to a wors-
drugs for secondary bacterial infections, and enteral nutrition. ened outcome.70 Administration of lysine tablets or paste could
Appetite stimulants such as diazepam may be useful in some be considered for client-owned cats with chronic herpesviral
cats, but if inappetence persists for longer than 3 days, feeding disease but is not recommended in shelter situations.
tube placement is indicated. Severely affected cats may require The use of glucocorticoids to treat refractory keratitis is con-
hospitalization in isolation and treatment with parenteral fluids, troversial, because it may reactivate virus and worsen disease.
nebulization, and supplemental oxygen. Acute signs generally However, eosinophilic keratitis may require treatment with top-
resolve within 2 to 3 weeks with supportive care, but some cats ical glucocorticoids. Because the role of FHV-1 in this condition
experience frequent disease relapses and chronic complications is unclear, topical antiviral medications such as cidofovir are
such as keratitis and persistent stomatitis. Cats with VSD may often given concurrently. Topical glucocorticoids are contrain-
require treatment with colloids. Mortality rates that exceed dicated when corneal ulceration is present, so the eyes should
50% can occur in outbreaks of VSD.31 first be carefully examined after staining with fluorescein. Cats
CHAPTER 23  Feline Respiratory Viral Infections 247

with ulcerative eosinophilic keratitis may require ­systemic glu- of virus by antibody), and the load of latent virus in the trigemi-
cocorticoid treatment until the ulcers heal. Referral to a veteri- nal ganglia.15,72,73 It is unclear if the frequency of reactivation is
nary ophthalmologist is recommended. reduced, although this is the case for herpesviral vaccines in other
species (such as the shingles vaccine for humans). The intranasal
Chronic Gingivostomatitis FHV-1 vaccine virus can itself become latent, but whether par-
Caudal stomatitis in cats infected with FCV is often refractory enteral FHV-1 vaccine virus becomes latent is unknown. Most
to medical treatment. Clinical cure occurs in 50% to 60% of FCV vaccines do not prevent shedding, and virus shedding from
cats and there is significant improvement in 30% to 40% of cats the oropharynx can actually increase after FCV vaccination,
after 1) extraction of teeth in the vicinity of lesions, 2) treat- because of shedding of attenuated live vaccine virus.
ment with antimicrobial drugs with activity against anaerobic Because FHV-1 strains are antigenically similar, the immune
and gram-positive aerobic bacteria, and 3) the use of antiseptic response to one FHV-1 strain cross protects against other FHV-1
mouthwashes (see Chapter 21). Cats that fail to respond to this strains. Greater variability in cross protection occurs for FCV
treatment may require long-term treatment with antimicrobial strains. Vaccines that contain F9, the most widely used FCV vac-
and antiinflammatory drugs, although one study showed mini- cine strain, provide partial protection against aerosol challenge
mal improvement in cats with refractory stomatitis that were with heterologous strains.74 Cross-neutralization studies have
treated with prednisolone.53 Recombinant feline IFN-ω has also shown that the antigenic composition of current field FCV strains
been used subcutaneously and orally to treat caudal stomati- may differ enough from the original F9 strain that the level of
tis. In a randomized, multicenter, controlled, double-blinded cross-protection induced by F9 vaccines may be suboptimal. As
study of cats with refractory caudal stomatitis and alveolar/ a result, it has been suggested that new FCV strains should be
buccal mucositis, oromucosal treatment with rfIFN-ω (0.1 mil- incorporated into FCV vaccines. However, vaccines that con-
lion units q24h) was associated with significant improvement in tain F9 still appear to generate cross-reactive antibody to a large
lesions and decreased pain scores from day 0 to day 90.53 number of currently circulating strains.75 A nonadjuvanted, inac-
tivated vaccine that contains two FCV strains (G1 and 431) is
Influenza Virus Infections available in Europe, which provided greater reduction in clini-
Oseltamivir has been used to treat captive wild and domestic cats cal scores after challenge with a heterologous strain that the use
with influenza virus infections,33,71 but it was not effective. The of either strain alone and also reduced shedding.76 Exposure of
optimal dose of this drug in cats is not known. Therefore, cats with vaccinated cats to field strains of FCV may also serve to boost
suspected influenza virus infections should be treated supportively. immune responses and ultimately broaden protection. Cats that
are not exposed to field strains, such as isolated indoor cats, may
Immunity and Vaccination be more susceptible to severe disease when hypervirulent strains
appear. An adjuvanted, inactivated vaccine for VSD that contains
Immunity to respiratory viruses depends on cell-mediated and a single hypervirulent strain was introduced in the United States
systemic and local (mucosal IgA) humoral immune responses. in 2007. However, the degree to which this vaccine cross-protects
Maternal antibody to respiratory viruses can persist until 14 against other hypervirulent strains is unknown, and outbreaks
weeks of age,15 although other kittens may be susceptible to cease when infection control measures are implemented. Because
infection at 6 weeks of age. of this and the increased risk of sarcoma formation with adju-
Vaccines for FHV-1 and FCV have been available for decades, vanted vaccines, the usefulness of the vaccine has been questioned.
but these do not provide complete protection, and disease con- Parenteral vaccines that contain FCV and FHV-1 are gener-
tinues to be widespread in the cat population. Infection of kittens ally safe, although mild signs of URTD have the potential to
before completion of the primary vaccine series contributes to this occur if the cat licks the site of vaccination or aerosolization
problem. Both inactivated and attenuated live FHV-1 and FCV of vaccine occurs during administration. Intranasal vaccines
vaccines are available, and attenuated live vaccines are available are less well tolerated by cats and can be occasionally be fol-
for parenteral or intranasal administration. Inactivated vaccines lowed by mild to moderate postvaccinal signs of URTD. This
contain adjuvant, and their use should be reserved for immu- may be problematic in shelter environments where clinical
nosuppressed cats (such as retrovirus-infected cats) or pregnant signs of URTD warrant the need for separate housing. On the
cats, if deemed necessary. When infections are a problem in kit- other hand, intranasal vaccines are advantageous in this situa-
tens in breeding catteries, vaccination of the queen before mating tion because of rapid onset of immunity, with partial protection
is preferable to vaccination during pregnancy and may prolong after 2 days and significant protection after 4 days.15 They can
the duration of maternal antibody persistence.50 Vaccination also overcome maternal antibody at a young age, although some
as early as 4 weeks of age (but not before) could be considered injectable vaccines may also still be effective at an early age.
in catteries with high rates of viral URTD. The primary series Because vaccine virus is propagated in feline kidney cells, there
should consist of vaccines administered every 3 to 4 weeks of age has been concern that parenteral administration of feline vac-
until no earlier than 16 weeks of age. cines might lead to chronic renal inflammation when immune
No respiratory viral vaccine prevents infection, the carrier responses to vaccine components cross-react with endogenous
state, or reactivation of FHV-1 infection with stress, but they renal antigens.77 Further studies are required to determine
can reduce disease severity. Whether a cat contracts feline viral whether this is clinically important.
URTD after vaccination depends on factors such as virus strain Vaccine boosters are recommended 1 year after completion
virulence, the challenge dose, environmental temperature and of the primary series, and at 3-yearly intervals thereafter in cats
humidity, and the presence of underlying immunosuppressive with low levels of exposure to respiratory viruses. For cats in
disease or other concurrent disease. contaminated environments such as catteries, boarding facili-
For FHV-1 infections, vaccination can reduce the duration of ties, or large multicat households with frequent introductions,
shedding, viral infectivity (presumably as a result of complexing annual vaccination may be more appropriate. The duration of
248 SECTION 1  Viral Diseases

immunity after vaccination remains incompletely understood. of FHV-1 or FCV infection, and cats examined for routine vac-
Although partial protection persists for several years after vac- cination purposes may be susceptible to nosocomial infection
cination,78 the degree of protection does wane with time, at with these viruses. Cats should be housed individually (unless
least for cats that receive no natural boostering in the interim they are from the same household and tolerate close proximity
(i.e., from exposure to field virus). In one report, relative FHV-1 to one another). Barriers between cats should be impermeable,
and FCV vaccine efficacies were 95% and 85%, respectively, and cats with URTD should be separated by at least 4 to 5 feet
when challenge occurred a few weeks after primary vaccination to prevent aerosol transmission. If possible, cats that enter large
with an inactivated vaccine.79 This compared with 52% and multicat environments and shelters should be quarantined for
63%, respectively, when challenge occurred after 7.5 years.80 3 weeks to identify cats that are incubating disease. When all
In another study, a 50% reduction in clinical scores occurred other control measures fail in breeding catteries, early weaning
after challenge 1 year after administration of an attenuated live and isolation of kittens from 4 weeks could be considered.
FHV-1 vaccine, compared with around 75% after challenge In regions where highly pathogenic H5N1 avian influenza
4 weeks after vaccination.81 Similar results were observed for virus infections have been identified in birds, cats should be kept
an inactivated FCV vaccine. In the field, natural boostering as indoors. Restriction zones defined for this purpose consist of an
a result of contact with virus shed by other cats or subclinical area within a 10-km radius of an outbreak of influenza in birds
reactivation may maintain immunity for longer periods of time. for 30 days after the discovery of infected birds.36 Cats should
also not be fed raw poultry. Separation of cats from avian spe-
Prevention cies in shelters and hospital situations should be maintained.

In addition to vaccination, reduction of stress and overcrowding Public Health Aspects


is critical for prevention of URTD, especially that due to FHV-
1. Proper hand washing and disinfection in shelters, boarding FHV-1 and FCV do not infect humans. Although cats can
facilities, and veterinary hospitals to prevent fomite transmis- acquire influenza virus infections from humans, the risk of
sion is also very important, and dedicated food and water bowls transmission from cats back to humans is unknown. Neverthe-
should be used. Inactivation of FCV requires the use of disin- less, cats suspected to have influenza virus infections (such as
fectants such as sodium hypochlorite (household bleach diluted during a recognized outbreak) should be isolated, the handling
1:30), potassium peroxymonosulfate (Trifectant, Virkon S), or of these cats should be minimized, and appropriate protective
accelerated hydrogen peroxide. Veterinarians should remember attire should be worn, which should ideally include properly
that all cats, whether apparently healthy or sick, can be a source fitted high-density surgical masks.

CASE EXAMPLE signs. He was last immunized for FHV-1, FCV, FPV, and rabies
1 year previously. Boris was housed completely indoors and
was exposed to one other cat that only occasionally had
Signalment: “Boris”, a 7-year-old male neutered Burmese cat access to the outdoors. His diet consisted of a commercial
from northern California. dry cat food.
History: Boris was brought to the University of California, Davis, Physical Examination:
Veterinary Medical Teaching Hospital for a 3-day history Body Weight: 4.8 kg
of inappetence, head-shaking, gagging, hypersalivation, General: Lethargic, but responsive, 5% to 7% dehydrated. Held
coughing, and sneezing. The gagging occurred throughout head extended and gagged and hypersalivated throughout
the day and occasionally was followed by vomiting. The the examination; seemed unable to swallow. Vomited bile-
vomit consisted of bile-stained fluid. Boris had not been stained fluid once during the examination. T = 100.8°F (38.2°C),
drinking. He was taken to a local emergency clinic where HR >240 beats/min, respiratory rate = 24 breaths/min, mucous
bilateral serous nasal discharge was noted, and he was membranes pink, CRT = 1 s. The cat’s haircoat was unkempt.
treated for 24 hours with intravenous crystalloid fluids and Eyes, Ears, Nose, and Throat: Bilateral serous nasal discharge
amoxicillin. Blood work showed hypokalemia (4.0 mEq/L, was present. A linear ulcer was identified on the right lateral
reference range 4.5 to 5.3 mEq/L), hyperglycemia (292 mg/ aspect of the tongue.
dL, reference range 70 to 150 mg/dL), neutrophilia (12,920 Musculoskeletal: Body condition score 4/9. Ambulatory.
cells/μL, reference range 2500 to 12,500) and lymphopenia Cardiovascular and Respiratory: Other than tachycardia, no
(136, reference range 1500 to 7000). FeLV and FIV serology other abnormalities were detected.
was negative. A urinalysis showed a specific gravity of Gastrointestinal and Genitourinary: Mild abdominal
1.050, pH of 7, and 1+ proteinuria and glucosuria. Lateral distention was present.
and dorsoventral radiographs of the thorax and abdomen Lymph Nodes: All lymph nodes were < 1 cm in diameter.
showed moderate to severe gas distention of the proximal Laboratory Findings:
esophagus and stomach. Profuse salivation and gagging CBC:
continued, and he was referred to a specialty clinic because HCT 34.2% (30%-50%)
of concerns for a possible gastrointestinal foreign body. MCV 51.0 fL (42-53 fL)
There had been no diarrhea, no history of travel or trauma, MCHC 32.5 g/dL (30-33.5 g/dL)
and no recent surgical procedures. A new indoor plant was WBC 12,600 cells/µL (4500-14,000 cells/µL)
brought into the house 2 days before the onset of clinical Neutrophils 11,844 cells/µL (2000-9000 cells/µL)
CHAPTER 23  Feline Respiratory Viral Infections 249

Band neutrophils 126 cells/µL twice daily. After the first day of hospitalization, the owners
Lymphocytes 126 cells/µL (1000-7000 cells/µL) reported that their other cat had developed signs of URTD,
Monocytes 252 cells/µL (50-600 cells/µL) with sneezing and nasal discharge. Enteral or parenteral
Eosinophils 126 cells/µL (150-1100 cells/µL) nutrition was discussed with the owner, but on the evening
Platelets 989,000 platelets/µL (180,000-500,000 platelets/ of the second day of hospitalization, Boris was found gagging
µL). The neutrophils showed slight toxicity, and there and cyanotic. He was taken to the intensive care unit but
were a few Heinz bodies developed cardiorespiratory arrest. CPR was instituted and
Serum Chemistry Profile: tracheal suction revealed yellow fluid consistent with possible
Sodium 141 mmol/L (151-158 mmol/L) aspiration. Although CPR was initially successful, Boris arrested
Potassium 4.0 mmol/L (3.6-4.9 mmol/L) again and died shortly afterward.
Chloride 108 mmol/L (117-126 mmol/L) Necropsy Examination: Gross necropsy examination
Bicarbonate 27 mmol/L (15-21 mmol/L) showed numerous lingual ulcerations, mostly pinpoint but
Phosphorus 4.5 mg/dL (3.2-6.3 mg/dL) some coalescing into larger ulcers up to 1 cm long and 0.5 cm
Calcium 8.1 mg/dL (9.0-10.9 mg/dl) wide. Both lungs were diffusely dark red, poorly collapsed,
BUN 21 mg/dL (18-33 mg/dL) and oozed large amounts of clear fluid on cut surface. There
Creatinine 1.1 mg/dL (1.1-2.2 mg/dL) was a small amount of yellow ascites fluid. Histopathology
Glucose 212 mg/dL (63-118 mg/dL) revealed marked, multifocal, acute to subacute necrotizing
Total protein 5.1 g/dL (6.6-8.4 g/dL) stomatitis, rhinitis, tracheitis, bronchiolitis, and alveolitis
Albumin 2.2 g/dL (2.2-4.6 g/dL) with intranuclear herpesviral inclusions in the bronchiolar
Globulin 2.9 g/dL (2.8-5.4 g/dL) epithelium. Immunofluorescent antibody testing on lung
ALT 83 U/L (27-101 U/L) tissue was positive for FHV-1. Virus isolation was negative.
AST 50 U/L (17-58 U/L) Histopathology of gastrointestinal biopsies collected
ALP 16 U/L (14-71 U/L) at endoscopy and tissues collected at necropsy was
Cholesterol 147 mg/dL (89-258 mg/dL) unremarkable.
Total bilirubin 0 mg/dL (0-0.2 mg/dL) Comments: This cat died of a severe and overwhelming
Endoscopy: Boris was anesthetized and an oral examination FHV-1 infection, despite the fact that he was an adult
performed, which showed only mild dorsal pharyngeal cat that had been vaccinated regularly. Clinical signs of
lymphoid hyperplasia. Rhinoscopy showed a moderate anorexia, gagging and hypersalivation likely resulted from
amount of mucus and mild diffuse erythema of the nasal stomatitis, pharyngitis, and tracheitis. Histopathology
mucosa. Gastroduodenoscopy revealed focal areas of gastric with immunofluorescent antibody staining confirmed the
erosion with hemorrhage. involvement of FHV-1. The negative virus isolation result
Treatment: Boris was treated with IV crystalloid fluids (LRS may have been because of an insufficient sample size,
with 20 mEq/L KCl at 20 mL/h), ampicillin (23 mg/kg IV inactivation of virus by antibody, or lack of viable virus
q8h), enrofloxacin (2.6 mg/kg IV q24h), sucralfate (0.25 g PO in the specimen. Because FHV-1 survives poorly in the
q8h), and famotidine (0.25 mg/kg IV q12h). He continued to environment, it may deteriorate in clinical specimens during
hypersalivate, and he vomited approximately 100 mL of fluid transport to the laboratory.

SUGGESTED READINGS 5. Helps CR, Lait P, Damhuis A, et al. Factors associated with upper
respiratory tract disease caused by feline herpesvirus, feline calicivi-
Ali A, Daniels JB, Zhang Y, et  al. Pandemic and seasonal human rus, Chlamydophila felis and Bordetella bronchiseptica in cats: expe-
influenza virus infections in domestic cats: Prevalence, association rience from 218 European catteries. Vet Rec. 2005;156:669-673.
with respiratory disease, and seasonality pattern. J Clin Microbiol. 6. Hamano M, Maeda K, Mizukoshi F, et al. Experimental infection
2011;49:4101-4105. of recent field isolates of feline herpesvirus type 1. J Vet Med Sci.
Gaskell R, Dawson S, Radford A, et  al. Feline herpesvirus. Vet Res. 2003;65:939-943.
2007;38:337-354. 7. Veir JK, Ruch-Gallie R, Spindel ME, et al. Prevalence of selected
Marschall J, Hartmann K. Avian influenza A H5N1 infections in cats. infectious organisms and comparison of two anatomic sampling
J Feline Med Surg. 2008;10:359-365. sites in shelter cats with upper respiratory tract disease. J Feline
Radford AD, Coyne KP, Dawson S, et  al. Feline calicivirus. Vet Res. Med Surg. 2008;10:551-557.
2007;38:319-335. 8. Bech-Nielsen S, Fulton RW, Cox HU, et al. Feline respiratory tract
disease in Louisiana. Am J Vet Res. 1980;41:1293-1298.
REFERENCES 9. Ellis TM. Feline respiratory virus carriers in clinically healthy cats.
1. Crandell RA, Maurer FD. Isolation of a feline virus associ- Aust Vet J. 1981;57:115-118.
ated with intranuclear inclusion bodies. Proc Soc Exp Biol Med. 10. Harbour DA, Howard PE, Gaskell RM. Isolation of feline calici-
1958;97:487-490. virus and feline herpesvirus from domestic cats 1980 to 1989. Vet
2. Fastier LB. A new feline virus isolated in tissue culture. Am J Vet Rec. 1991;128:77-80.
Res. 1957;18:382-389. 11. Shewen PE, Povey RC, Wilson MR. A survey of the conjunctival
3. Songserm T, Amonsin A, Jam-on R, et al. Avian influenza H5N1 in flora of clinically normal cats and cats with conjunctivitis. Can Vet J.
naturally infected domestic cat. Emerg Infect Dis. 2006;12:681-683. 1980;21:231-233.
4. Sponseller BA, Strait E, Jergens A, et  al. Influenza A pandemic 12. Wardley RC, Gaskell RM, Povey RC. Feline respiratory viruses—
(H1N1) 2009 virus infection in domestic cat. Emerg Infect Dis. their prevalence in clinically healthy cats. J Small Anim Pract.
2010;16:534-537. 1974;15:579-586.
250 SECTION 1  Viral Diseases

13. Coutts AJ, Dawson S, Willoughby K, et al. Isolation of feline respi- 37. Kuiken T, Rimmelzwaan G, van Riel D, et al. Avian H5N1 influ-
ratory viruses from clinically healthy cats at UK cat shows. Vet Rec. enza in cats. Science. 2004;306:241.
1994;135:555-556. 38. Rimmelzwaan GF, van Riel D, Baars M, et  al. Influenza A virus
14. Ruch-Gallie RA, Veir JK, Hawley JR, et  al. Results of molecular (H5N1) infection in cats causes systemic disease with potential
diagnostic assays targeting feline herpesvirus-1 and feline calicivi- novel routes of virus spread within and between hosts. Am J Pathol.
rus in adult cats administered modified live vaccines. J Feline Med 2006;168:176-183;quiz 364.
Surg. 2011;13:541-545. 39. Klopfleisch R, Wolf PU, Uhl W, et al. Distribution of lesions and
15. Gaskell R, Dawson S, Radford A, et al. Feline herpesvirus. Vet Res. antigen of highly pathogenic avian influenza virus A/Swan/Ger-
2007;38:337-354. many/R65/06 (H5N1) in domestic cats after presumptive infection
16. Gaskell RM, Povey RC. Re-excretion of feline viral rhinotracheitis by wild birds. Vet Pathol. 2007;44:261-268.
virus following corticosteroid treatment. Vet Rec. 1973;93:204-205. 40. Leschnik M, Weikel J, Mostl K, et  al. Subclinical infection
17. Gaskell RM, Povey RC. Experimental induction of feline viral with avian influenza A (H5N1) virus in cats. Emerg Infect Dis.
rhinotracheitis virus re-excretion in FVR-recovered cats. Vet Rec. 2007;13:243-247.
1977;100:128-133. 41. Marschall J, Schulz B, Harder Priv-Doz TC, et al. Prevalence of influ-
18. Gaskell RM, Povey RC. Transmission of feline viral rhinotrache- enza A H5N1 virus in cats from areas with occurrence of highly patho-
itis. Vet Rec. 1982;111:359-362. genic avian influenza in birds. J Feline Med Surg. 2008;10:355-358.
19. Di Martino B, Di Rocco C, Ceci C, et  al. Characterization of a 42. Song DS, An DJ, Moon HJ, et al. Interspecies transmission of the
strain of feline calicivirus isolated from a dog faecal sample. Vet canine influenza H3N2 virus to domestic cats in South Korea,
Microbiol. 2009;139:52-57. 2010. J Gen Virol. 2011;92:2350-2355.
20. Radford AD, Turner PC, Bennett M, et al. Quasispecies evolution 43. Harder TC, Vahlenkamp TW. Influenza virus infections in dogs
of a hypervariable region of the feline calicivirus capsid gene in and cats. Vet Immunol Immunopathol. 2010;134:54-60.
cell culture and in persistently infected cats. J Gen Virol. 1998;79 44. van Riel D, Rimmelzwaan GF, van Amerongen G, et  al. Highly
(Pt 1):1-10. pathogenic avian influenza virus H7N7 isolated from a fatal human
21. Wardley RC. Feline calicivirus carrier state. A study of the host/ case causes respiratory disease in cats but does not spread systemi-
virus relationship. Arch Virol. 1976;52:243-249. cally. Am J Pathol. 2010;177:2185-2190.
22. Di Martino B, Ceci C, Di Profio F, et  al. In  vitro inactivation of 45. Ali A, Daniels JB, Zhang Y, et al. Pandemic and seasonal human
feline calicivirus (FCV) by chemical disinfectants: resistance varia- influenza virus infections in domestic cats: Prevalence, association
tion among field strains. Arch Virol. 2010;155:2047-2051. with respiratory disease, and seasonality pattern. J Clin Microbiol.
23. Doultree JC, Druce JD, Birch CJ, et al. Inactivation of feline calici- 2011;49:4101-4105.
virus, a Norwalk virus surrogate. J Hosp Infect. 1999;41:51-57. 46. Nasisse MP, Guy JS, Davidson MG, et  al. Experimental ocular
24. Smid B, Valicek L, Rodak L, et al. Rabbit haemorrhagic disease: an herpesvirus infection in the cat. Sites of virus replication, clinical
investigation of some properties of the virus and evaluation of an features and effects of corticosteroid administration. Invest Oph-
inactivated vaccine. Vet Microbiol. 1991;26:77-85. thalmol Vis Sci. 1989;30:1758-1768.
25. Mencke N, Vobis M, Mehlhorn H, et  al. Transmission of feline 47. van den Brand JM, Stittelaar KJ, van Amerongen G, et al. Experi-
calicivirus via the cat flea (Ctenocephalides felis). Parasitol Res. mental pandemic (H1N1) 2009 virus infection of cats. Emerg Infect
2009;105:185-189. Dis. 2010;16:1745-1747.
26. Hurley KE, Pesavento PA, Pedersen NC, et  al. An outbreak of 48. Volopich S, Benetka V, Schwendenwein I, et al. Cytologic findings,
virulent systemic feline calicivirus disease. J Am Vet Med Assoc. and feline herpesvirus DNA and Chlamydophila felis antigen detec-
2004;224:241-249. tion rates in normal cats and cats with conjunctival and corneal
27. Pedersen NC, Elliott JB, Glasgow A, et al. An isolated epizootic of lesions. Vet Ophthalmol. 2005;8:25-32.
hemorrhagic-like fever in cats caused by a novel and highly virulent 49. Sanchez MD, Goldschmidt MH, Mauldin EA. Herpesvirus derma-
strain of feline calicivirus. Vet Microbiol. 2000;73:281-300. titis in two cats without facial lesions. Vet Dermatol. 2012;23:171-
28. Reynolds BS, Poulet H, Pingret JL, et  al. A nosocomial outbreak 173, e135.
of feline calicivirus associated virulent systemic disease in France. 50. Radford AD, Addie D, Belak S, et  al. Feline calicivirus infection.
J Feline Med Surg. 2009;11:633-644. ABCD guidelines on prevention and management. J Feline Med
29. Schulz BS, Hartmann K, Unterer S, et al. Two outbreaks of virulent Surg. 2009;11:556-564.
systemic feline calicivirus infection in cats in Germany. Berl Munch 51. Belgard S, Truyen U, Thibault JC, et al. Relevance of feline calici-
Tierarztl Wochenschr. 2011;124:186-193. virus, feline immunodeficiency virus, feline leukemia virus, feline
30. Radford AD, Gaskell RM. Dealing with a potential case of FCV- herpesvirus and Bartonella henselae in cats with chronic gingivo-
associated virulent systemic disease. Vet Rec. 2011;168:585-586. stomatitis. Berl Munch Tierarztl Wochenschr. 2010;123:369-376.
31. Coyne KP, Jones BR, Kipar A, et  al. Lethal outbreak of dis- 52. Dowers KL, Hawley JR, Brewer MM, et al. Association of Barton-
ease associated with feline calicivirus infection in cats. Vet Rec. ella species, feline calicivirus, and feline herpesvirus 1 infection with
2006;158:544-550. gingivostomatitis in cats. J Feline Med Surg. 2010;12:314-321.
32. Meyer A, Kershaw O, Klopfleisch R. Feline calicivirus-associated 53. Hennet PR, Camy GA, McGahie DM, et al. Comparative efficacy
virulent systemic disease: not necessarily a local epizootic problem. of a recombinant feline interferon omega in refractory cases of cali-
Vet Rec. 2011;168:589. civirus-positive cats with caudal stomatitis: a randomised, multi-
33. Lohr CV, DeBess EE, Baker RJ, et al. Pathology and viral antigen centre, controlled, double-blind study in 39 cats. J Feline Med Surg.
distribution of lethal pneumonia in domestic cats due to pandemic 2011;13:577-587.
(H1N1) 2009 influenza A virus. Vet Pathol. 2010;47:378-386. 54. Pesavento PA, Stokol T, Liu H, et al. Distribution of the feline cali-
34. Fiorentini L, Taddei R, Moreno A, et  al. Influenza A pandemic civirus receptor junctional adhesion molecule A in feline tissues.
(H1N1) 2009 virus outbreak in a cat colony in Italy. Zoonoses Vet Pathol. 2011;48:361-368.
Public Health. 2011;58:573-581. 55. Lee M, Bosward KL, Norris JM. Immunohistological evaluation of
35. Ali A, Daniels JB, Zhang Y, et al. Pandemic and seasonal human feline herpesvirus-1 infection in feline eosinophilic dermatoses or
influenza virus infections in domestic cats: prevalence, association stomatitis. J Feline Med Surg. 2010;12:72-79.
with respiratory disease, and seasonality patterns. J Clin Microbiol. 56. Marschall J, Schulz B, Hartmann K. Evaluation of a point-of-
2011;49:4101-4105. care influenza antigen test for the detection of highly pathogenic
36. Marschall J, Hartmann K. Avian influenza A H5N1 infections in avian influenza H5N1 virus in cats. Transbound Emerg Dis.
cats. J Feline Med Surg. 2008;10:359-365. 2008;55:315-317.
CHAPTER 23  Feline Respiratory Viral Infections 251

57. Crawford C, Spindel M. Canine influenza. In: Miller L, Hurley K, 69. Rees TM, Lubinski JL. Oral supplementation with l-lysine did not
eds. Infectious Disease Management in Animal Shelters. Ames, IA: prevent upper respiratory infection in a shelter population of cats.
Wiley-Blackwell; 2009:173-180. J Feline Med Surg. 2008;10:510-513.
58. Storey ES, Gerding PA, Scherba G, et  al. Survival of equine her- 70. Drazenovich TL, Fascetti AJ, Westermeyer HD, et  al. Effects of
pesvirus-4, feline herpesvirus-1, and feline calicivirus in multidose dietary lysine supplementation on upper respiratory and ocular dis-
ophthalmic solutions. Vet Ophthalmol. 2002;5:263-267. ease and detection of infectious organisms in cats within an animal
59. Burgesser KM, Hotaling S, Schiebel A, et al. Comparison of PCR, shelter. Am J Vet Res. 2009;70(11):1391-1400.
virus isolation, and indirect fluorescent antibody staining in the 71. Thanawongnuwech R, Amonsin A, Tantilertcharoen R, et al. Prob-
detection of naturally occurring feline herpesvirus infections. J Vet able tiger-to-tiger transmission of avian influenza H5N1. Emerg
Diagn Invest. 1999;11:122-126. Infect Dis. 2005;11:699-701.
60. Goldschmidt P, Rostane H, Saint-Jean C, et  al. Effects of topical 72. Sykes JE, Browning GF, Anderson G, et al. Differential sensitivity
anaesthetics and fluorescein on the real-time PCR used for the diag- of culture and the polymerase chain reaction for detection of feline
nosis of herpesviruses and Acanthamoeba keratitis. Br J Ophthal- herpesvirus 1 in vaccinated and unvaccinated cats. Arch Virol.
mol. 2006;90:1354-1356. 1997;142:65-74.
61. Segarra S, Papasouliotis K, Helps C. The in  vitro effects of 73. Sussman MD, Maes RK, Kruger JM. Vaccination of cats for
proxymetacaine, fluorescein, and fusidic acid on real-time PCR feline rhinotracheitis results in a quantitative reduction of viru-
assays used for the diagnosis of feline herpesvirus-1 and Chla- lent feline herpesvirus-1 latency load after challenge. Virology.
mydophila felis infections. Vet Ophthalmol. 2011;14(Suppl. 1997;228:379-382.
1):5-8. 74. Kahn DE, Hoover EA, Bittle JL. Induction of immunity to feline
62. Burns RE, Wagner DC, Leutenegger CM, et  al. Histologic and caliciviral disease. Infect Immun. 1975;11:1003-1009.
molecular correlation in shelter cats with acute upper respiratory 75. Porter CJ, Radford AD, Gaskell RM, et al. Comparison of the abil-
infection. J Clin Microbiol. 2011;49:2454-2460. ity of feline calicivirus (FCV) vaccines to neutralise a panel of cur-
63. Malik R, Lessels NS, Webb S, et al. Treatment of feline herpesvi- rent UK FCV isolates. J Feline Med Surg. 2008;10:32-40.
rus-1 associated disease in cats with famciclovir and related drugs. 76. Poulet H, Brunet S, Leroy V, et al. Immunisation with a combina-
J Feline Med Surg. 2009;11:40-48. tion of two complementary feline calicivirus strains induces a broad
64. Thomasy SM, Lim CC, Reilly CM, et  al. Evaluation of orally cross-protection against heterologous challenges. Vet Microbiol.
administered famciclovir in cats experimentally infected with feline 2005;106:17-31.
herpesvirus type-1. Am J Vet Res. 2011;72:85-95. 77. Whittemore JC, Hawley JR, Jensen WA, et  al. Antibodies against
64a. Lim CC, Reilly CM, Thomasy SM, et al. Effects of feline herpesvi- Crandell Rees feline kidney (CRFK) cell line antigens, alpha-enolase,
rus type 1 on tear film break-up time, Schirmer tear test results, and and annexin A2 in vaccinated and CRFK hyperinoculated cats. J Vet
conjunctival goblet cell density in experimentally infected cats. Am Intern Med. 2010;24:306-313.
J Vet Res. 2009;70:394-403. 78. Gore TC, Lakshmanan N, Williams JR, et al. Three-year duration
65. Fontenelle JP, Powell CC, Veir JK, et  al. Effect of topical oph- of immunity in cats following vaccination against feline rhinotra-
thalmic application of cidofovir on experimentally induced pri- cheitis virus, feline calicivirus, and feline panleukopenia virus. Vet
mary ocular feline herpesvirus-1 infection in cats. Am J Vet Res. Ther. 2006;7:213-222.
2008;69:289-293. 79. Povey RC, Koonse H, Hays MB. Immunogenicity and safety of
66. Cocker FM, Howard PE, Harbour DA. Effect of human alpha- an inactivated vaccine for the prevention of rhinotracheitis, cali-
hybrid interferon on the course of feline viral rhinotracheitis. Vet civiral disease, and panleukopenia in cats. J Am Vet Med Assoc.
Rec. 1987;120:391-393. 1980;177:347-350.
67. Stiles J, Townsend WM, Rogers QR, et al. Effect of oral adminis- 80. Scott FW, Geissinger CM. Long-term immunity in cats vaccinated
tration of l-lysine on conjunctivitis caused by feline herpesvirus in with an inactivated trivalent vaccine. Am J Vet Res. 1999;60:652-658.
cats. Am J Vet Res. 2002;63:99-103. 81. Poulet H. Alternative early life vaccination programs for compan-
68. Maggs DJ, Nasisse MP, Kass PH. Efficacy of oral supplementation ion animals. J Comp Pathol. 2007;137(Suppl. 1):S67-S71.
with l-lysine in cats latently infected with feline herpesvirus. Am J
Vet Res. 2003;64:37-42.
CHAPTER 24

Feline Poxvirus Infections


Malcolm Bennett

a range of independent virus species is still to be determined.


Overview of Feline Poxvirus Infections For the purpose of this account, however, they are regarded as
First Described: Feline poxvirus infection was first described a single viral species.
in 19781 Despite its name, cowpox is rarely described in cattle; cow-
Causes: Poxviruses (family Poxviridae). Cowpox virus infec- pox virus circulates primarily in wild rodents. It has a wide
tion is by far the most frequently reported poxviral infec- host range that includes not only rodents, cats, dogs, cattle,
tion of both cats and dogs. Other poxvirus infections, and human beings, but a variety of captive wildlife mam-
including parapoxvirus (assumed to be orf virus) and mals. The main route of transmission among rodents is not
raccoonpox virus infections, have also been described known, although, as for many orthopoxviruses, the respira-
in pets. Other poxviruses are also likely to infect cats and tory route is probably important. In most accidental hosts,
dogs but go unreported. which include domestic animals and human beings, the most
frequent source of infection is through a break in the skin.
Affected Hosts: Cats, less often dogs, and a variety of other
This gives rise to the “primary” lesion. The main reservoirs of
mammalian host species
cowpox virus are voles and, to a slightly lesser extent, Apode-
Geographic Distribution: Cowpox virus is limited to Europe mus mice. As these rodent species undergo annual population
and Asia. Infections with other poxviruses have been cycles, breeding mainly in the summer and fall months, viral
described in North America. transmission and prevalence has strong seasonality. Transmis-
Mode of Transmission: Entry through a break in the skin sion to accidental hosts such as cats, dogs, people, and zoo
animals therefore also occurs mainly (but not exclusively) in
Major Clinical Signs: Single or multiple, papular, and crusted the late summer, fall, and early winter. Cats probably become
skin lesions. Fever, lethargy, inappetence, and signs of infected directly from rodent hunting, whereas infection in
pneumonia can also occur in affected cats. other species is often acquired from a “liaison” or “amplify-
Differential Diagnoses: Allergic dermatitis, facial dermatitis ing” host. These are accidental hosts in which the virus rep-
caused by feline herpesvirus-1, nocardiosis, mycobacte- licates to higher titers than in the reservoir rodent hosts; thus
riosis, cutaneous lymphoma, autoimmune dermatoses they bridge the epidemiologic gap between the reservoir and
such as pemphigus foliaceus, cutaneous drug reactions the host of interest. The main liaison hosts for human cowpox
Human Health Significance: Many poxviruses, including cow- are the domestic cat (and, in the past, possibly cattle), fol-
pox and orf viruses, are zoonotic, and owners of affected lowed by peridomestic rodent species such as rats. Rats can
animals need to be advised of the threat to their health. also be a source of infection for captive wildlife species. The
virus is very hardy and survives in scabs for weeks or months,
including in the environment, where it may be a source of
human infection.
Etiology and Epidemiology
Raccoonpox Virus
Cowpox Virus Raccoonpox virus is also an orthopoxvirus. Remarkably little
Cowpox virus is a member of the Orthopoxvirus genus within is known about its epidemiology, or indeed about the other
the family Poxviridae, which are large, enveloped, double- known North American orthopoxviruses, skunkpox, and
stranded DNA viruses.2 Other orthopoxviruses include the volepox viruses, which together form a distinct clade. Raccoon-
viruses of smallpox (now eradicated), vaccinia (the smallpox pox virus appears to be fairly host specific but was isolated from
vaccine), monkeypox (which is endemic to central Africa but a cat with cowpox-like disease in Canada.3
was recently introduced to, and eradicated from, North Amer- Raccoonpox virus was originally isolated from the lungs of
ica), and a range of other mammalian orthopoxviruses. All apparently healthy raccoons in the northeastern United States,
orthopoxviruses are closely related genetically and antigenically; and serologic surveys in the same area suggested that more than
therefore, vaccinia virus can be used to immunize against all 20% of wild raccoons had antibody to the virus. Experimen-
orthopoxvirus infections. Cowpox virus has the largest genome tal infection of raccoons also caused no obvious disease.4 The
of all the orthopoxviruses. Different strains of cowpox virus narrow host range, low zoonotic and pathogenic potential, and
exist, as determined by both biologic characteristics and nucleo- endemicity of raccoonpox virus to North America has led to
tide sequence. Whether these merely reflect geographic variation its development as a vector for recombinant vaccines for use in
of the virus (and therefore its main hosts and pathogenicity) or American wildlife species.

252
CHAPTER 24  Feline Poxvirus Infections 253

Parapoxviruses
The parapoxviruses comprise a different genus from the
orthopoxviruses in the family Poxviridae. Parapoxviruses are
genetically very similar to each other (but significantly differ-
ent antigenically and genetically from the orthopoxviruses), and
differentiation of individual species is difficult.5 Parapoxvirus
infections have been described in cats in several countries, and
when characterized, the causative virus has been shown to be
orf virus.6 Orf occurs worldwide, mainly in sheep and goats,
and is readily transmissible to human beings.
Parapoxviruses, like many other poxviruses, survive in the
environment for long periods—possibly months or even years
under the right conditions. So although cats can become infected
through direct contact with sheep or goats, it is more likely that
infection occurs through contact with a contaminated fomite—
perhaps a piece of barbed wire or fencing that contacts a preex-
isting wound. The rarity of parapoxvirus infection in cats makes
its study very difficult.

Clinical Features FIGURE 24-1  Secondary papules and crusted skin lesions in a cat with ­cowpox virus
infection.
Signs and Their Pathogenesis
Cowpox Virus and decreased survival.10 When lesions are seen in rodents, they
In many hosts, such as human beings, dogs, and cattle, the pri- are often in “accidental” rodent hosts such as rats.
mary lesion of cowpox virus infection, plus perhaps some virus
replication in draining lymph nodes, may be as far as disease Other Poxviruses
progresses. In cats and many captive wild mammalian species, Raccoonpox has only been reported once in a domestic cat,3 in
however, the virus replicates in monocytes and macrophages which it caused a disease that resembled the primary lesion of
and spreads to multiple organs, particularly the lungs and the cowpox. The cat was initially evaluated for a swollen, crusty
spleen. Here, the virus replicates further, giving rise to a second- lesion on a thoracic limb digit, and 4 weeks later infection of the
ary viremia, which is often associated with pyrexia and mild or neighboring digit developed. Widespread secondary lesions did
occasionally severe systemic signs. Pneumonia can itself be fatal, not develop, and the cat recovered completely. Human infection
but usually the secondary viremia leads to a “secondary rash” with a raccoonpox recombinant vaccine led to a single lesion
of lesions distributed across the skin and mucosae. on the inoculated finger, which became swollen and painful
The signs of cowpox vary according to the host species. The and was accompanied by axillary lymphadenopathy. Complete
classical presentation in cats is of a primary skin lesion, often with recovery occurred within 4 weeks.
secondary bacterial infection, usually on a forelimb or the face, Parapoxvirus infections have been described in cats in the
thought to arise from the inoculation of virus into a wound— United Kingdom and New Zealand.5,10 The British cat devel-
perhaps a bite wound—while hunting infected wild rodents. In oped multiple crusted lesions on its face and dorsum that healed
some cases, severe necrotizing facial dermatitis develops. The pri- within a few weeks. Two cats from farms in New Zealand had
mary lesion may be followed by lethargy and possibly pyrexia for more severe clinical signs. One had a recurrent lesion on the
up to 10 days, after which multifocal skin lesions appear. These paw of a pelvic limb. The other had a nonhealing lesion on the
first develop as papules (Figure 24-1), which become ulcers up to paw of a thoracic limb that progressed, despite surgery, over
1-cm in diameter that crust over and heal over 4 to 6 weeks. This four months. Both cats were euthanized.
leaves small patches of alopecia until the hair regrows. In cats
such as Siamese, the hair can grow back a darker color, and scars Diagnosis
are often visible for years in cats if their hair is clipped. Infection
of dogs is less common, and in dogs, lesions are usually more Definitive diagnosis of poxvirus infections requires laboratory
localized and the systemic disease much less severe.6-8 identification of the causative virus. In Europe, where cowpox
In many captive wild Felidae, pneumonia is more com- virus is endemic, clinical signs combined with identification of
mon than in domestic cats, and mortality is high. Similarly, an orthopoxvirus probably suffice to make a diagnosis. Else-
elephants and rhinoceroses in central European zoos develop where, however, more extensive laboratory tests are required to
severe systemic disease and often die—indeed, this can be such identify the causative virus more precisely.
a severe disease that many European zoological parks vaccinate
their elephants. There are few reports of fatal cowpox in cap- Microbiologic Tests
tive primates,9 but there are anecdotal reports of high rates of Diagnostic assays available for poxvirus infections are shown
subclinical seroconversion in other primate populations. Other in Table 24-1.
affected zoo-kept species include giant anteater, beavers, okapi,
banded mongoose, jaguarondi, and the cavy. In wild rodent Cell Culture
reservoir hosts, clinical signs are absent or, at least, subtle. The best specimens to collect for virus isolation are fresh skin
Population-level studies of naturally infected voles and mice lesions, although virus can be found in crusts from healing
show that infection may be associated with decreased fecundity lesions for several weeks.11 No special transport media are
254 SECTION 1  Viral Diseases

TABLE 24-1
Diagnostic Assays Available for Poxvirus Infections
Assay Specimen Type Target Performance
Virus isolation Skin biopsies, crusts Poxviruses May not be widely available in some regions. Sensitive for di-
agnosis of orthopoxviruses, but parapoxviruses are difficult
to culture.
Polymerase chain Skin biopsies, crusts Poxvirus DNA Sensitivity and specificity may vary depending on assay design;
reaction (PCR) assays may not differentiate between orthopoxviruses.
Availability on a commercial basis may be limited in some
regions. Assays for parapoxviruses are not widely available.
Histopathology Skin biopsies, nec- Poxvirus inclusions Histopathology is similar for cowpox virus and raccoonpox
ropsy specimens virus. Confirmation of poxvirus infection requires immuno-
histochemistry or PCR.
Electron micros- Skin crusts Virus particles May not be widely available, turnaround time can be slow,
copy and may be expensive. Orthopoxviruses and parapoxviruses
can be differentiated based on their morphology.

needed. Lesions can be placed into a dry container for shipping


to the laboratory at room temperature. The virus replicates in
a variety of cell lines, in which it causes an obvious cytopathic
effect (CPE) that resembles that of feline herpesvirus-1. Unlike
most feline viruses, however, cowpox virus grows easily in cell
lines derived from other mammalian species, including primates.
Lesion material is ground or finely chopped and freeze-thawed
several times to release virus particles. CPE usually appears
within 24 to 76 hours and comprises plaques of syncytia fol-
lowed by rounding and loss of cells from the center of the
plaque. Staining with hematoxylin and eosin reveals large, intra-
cytoplasmic, eosinophilic inclusion bodies. Further confirmation
can be by immunostaining (e.g., immunofluorescence) to iden-
tify the virus as an orthopoxvirus, or PCR assay and sequencing.
Although raccoonpox virus grows in a variety of cell lines,
diagnosis is more likely to be made by histopathology and PCR
assays. This is because the virus is unlikely to be suspected ini-
tially, and biopsy and histology would be the first response of
most veterinarians. Parapoxviruses are notoriously difficult to
grow in cell culture, requiring primary cell cultures.

Molecular Diagnosis Using the Polymerase Chain Reaction


Some diagnostic laboratories offer PCR assays for diagnosis
of poxvirus infection, which can be both sensitive and, if com-
bined with sequencing, specific. Orthopoxviruses are so similar FIGURE 24-2  Electron micrograph of a cowpox virus particle. In contrast, parapoxvi-
genetically that PCRs targeted at many orthopoxvirus genes ruses have the appearance of a new ball of yarn, with threadlike surface tubules arranged
only identify orthopoxviruses to the genus level. PCRs that dif- in a crisscross fashion.
ferentiate between orthopoxvirus species have been described,
but such assays tend to be available through research or other orthopoxvirus particles to be seen in most cases (Figure 24-2).
specialist laboratories. Of course, in much of Europe, cowpox The diagnosis of parapoxvirus infection usually relies on elec-
virus is the only orthopoxvirus known to infect cats and dogs, so tron microscopy and visualization of the characteristic parapox-
identification to the species level is often not necessary. Unlike virus morphology, which resembles a “ball of yarn.”
orthopoxviruses, for which worries over bioterrorism have led
to the development of several sensitive and specific PCR assays, Serologic Diagnosis
molecular assays for parapoxviruses are not widely available. Previous cowpox virus infection can be diagnosed by various
serologic assays including immunofluorescent antibody, ELISA,
Direct Electron Microscopy and virus neutralization assays. The prevalence of orthopoxvi-
Electron microscopy of skin lesions is a more rapid approach rus antibodies in domestic cats in Europe is usually only 1% to
to diagnosis than cell culture and is fairly sensitive. Nega- 2%, so a consistent clinical history combined with an antibody
tive-stained suspensions of skin lesion material allow typical titer is reasonably specific.
CHAPTER 24  Feline Poxvirus Infections 255

Treatment and Prognosis


Treatment of poxvirus infections is supportive. There are no spe-
cific antiviral agents available. Most cats with cowpox recover
completely, but some, particularly if immunosuppressed by
concurrent infection or treatment with, for example, glucocorti-
coids, develop larger, persistent skin lesions, and/or pneumonia.
Cats that develop pneumonia often die despite supportive care.

Prevention
Housing cats indoors should help to prevent poxvirus infection
in cats. No vaccine is available. However, canarypox-vectored
vaccines are used to prevent rabies virus and FeLV infection in
cats and distemper in dogs.

Public Health Aspects


Cowpox virus infection in humans is usually limited to a skin
lesion at the site of inoculation (often on the hand or face)
accompanied by local lymphadenopathy and flu-like symp-
toms. The lesions can be painful, and an important differential
diagnosis is cutaneous anthrax. In Europe, most human cases
can be traced to contact with an infected cat; infection from
FIGURE 24-3  Histopathology of a feline poxvirus lesion that shows epithelial hyper- rodents or dogs is a much less frequent event. Although most
plasia and large numbers of eosinophilic intracytoplasmic inclusion bodies. patients recover uneventfully, immunosuppression can lead to
more extensive lesions and, rarely, even death.12,14-16 Although
thought not to be zoonotic, a recombinant raccoonpox vaccine
caused lesions in a laboratory worker.17 Orf is readily transmis-
Pathologic Findings sible to humans, usually from contact with sheep and goats.
Many cases of cowpox in cats, dogs, and humans are diagnosed Although transmission from an infected cat to a human being
by histopathology.6,12,13 The histology of cowpox is quite char- has not been reported, human orf associated with a cat scratch
acteristic; the virus causes hyperplasia and hypertrophy of the was reported from Utah in the United States.13
epithelium, with infected cells often containing eosinophilic,
intracytoplasmic inclusions that can be readily observed in SUGGESTED READINGS
routine hematoxylin and eosin–stained sections (Figure 24-3). Fairley RA, Whelan EM, Pesavento PA, et al. Recurrent localised cuta-
These A-type inclusions comprise virus-encoded protein that neous parapoxvirus infection in three cats. N Z Vet J. 2008;56:
forms a protective mass around newly formed virus particles. 196-201.
As the epithelium thickens, cells in the middle layers lyse, which Glatz M, Richter S, Ginter-Hanselmayer G, et al. Human cowpox in a
gives rise to fluid-filled lacunae that, when large enough, can veterinary student. Lancet Infect Dis. 2010;10:288.
Yager JA, Hutchison L, Barrett JW. Raccoonpox in a Canadian cat. Vet
be seen with the naked eye as the vesicles (or pocks/pox) from
Dermatol. 2006;17:443-448.
which these viruses get their name. Usually the vesicles quickly
ulcerate in cats. Older lesions undergo secondary bacterial REFERENCES
infection and subsequent neutrophil infiltration before heal-
1. Thomsett LR, Baxby D, Denham EM. Cowpox in the domestic cat.
ing. Immunostaining is useful in histopathologic diagnosis, Vet Rec. 1978;103:567.
especially in older lesions or those with secondary bacterial 2. Bennett M, Smith GL, Baxby D. Cowpox virus. In: Mahy BWJ,
infection. Regenmortel MHVV, eds. Encyclopedia of Virology. 3rd ed.
The histopathology of raccoonpox resembles that of Oxford: Academic Press; 2008.
cowpox.3 Hyperplastic, hypertrophic epithelium is present 3. Yager JA, Hutchison L, Barrett JW. Raccoonpox in a Canadian cat.
with eosinophilic, intracytoplasmic A-type inclusion bodies. Vet Dermatol. 2006;17:443-448.
Because North American orthopoxviruses are quite distinct 4. Thomas EK, Palmer EL, Obijeski JF, et al. Further characterization
from “old world” (Eurasian) orthopoxviruses, PCR assays and of Raccoonpox virus. Arch Virol. 1975;49:217-227.
sequencing can readily characterize the virus, even from fixed 5. Fairley RA, Whelan EM, Pesavento PA, et al. Recurrent localised
cutaneous parapoxvirus infection in three cats. N Z Vet J.
material.11
2008;56:196-201.
Histologic examination of parapoxvirus lesions from the 6. Bennett M, Gaskell CJ, Gaskell RM, et al. Poxvirus infection in the
New Zealand cats revealed epithelial hyperplasia giving rise to domestic cat: some clinical and epidemiological observations. Vet
multiple papillary projections from the skin surface, covered in Rec. 1986;118:387-390.
a thick layer of compacted keratin.5 Orf virus does not pro- 7. Kaysser P, von Bomhard W, Dobrzykowski L, et al. Genetic diver-
duce the A-type inclusions often seen with orthopoxviruses, but sity of feline cowpox virus, Germany 2000-2008. Vet Microbiol.
occasional basophilic or amphophilic inclusion bodies may be 2010;141:282-288.
present (so-called B-type inclusions, which consist of condensed 8. von Bomhard W, Mauldin EA, Breuer W, et al. Localized cowpox infec-
areas of virus replication). tion in a 5-month-old Rottweiler. Vet Dermatol. 2011;22:111-114.
256 SECTION 1  Viral Diseases

9. Girling SJ, Pizzi R, Cox A, et al. Fatal cowpox virus infection in 14. Baxby D, Bennett M. Poxvirus zoonoses. J Med Microbiol.
two squirrel monkeys (Saimiri sciureus). Vet Rec. 2011;169:156. 1997;46:17-20:28-33.
10. Hamblet CN. Parapoxvirus in a cat. Vet Rec. 1993;132:144. 15. Favier AL, Flusin O, Lepreux S, et al. Necrotic ulcerated lesion in
11. Bennett M, Baxby D, Gaskell RM, et al. The laboratory diagnosis a young boy caused by cowpox virus infection. Case Rep Dermatol.
of Orthopoxvirus infection in the domestic cat. J Small Anim Pract. 2011;3:186-194.
1985;26:653-661. 16. Glatz M, Richter S, Ginter-Hanselmayer G, et al. Human cowpox
12. Baxby D, Bennett M, Getty B. Human cowpox 1969-93: a review in a veterinary student. Lancet Infect Dis. 2010;10:288.
based on 54 cases. Br J Dermatol. 1994;131:598-607. 17. Rocke TE, Dein FJ, Fuchsberger M, et al. Limited infection upon
13. Frandsen J, Enslow M, Bowen AR. Orf parapoxvirus infection human exposure to a recombinant raccoon pox vaccine vector.
from a cat scratch. Dermatol Online J. 2011;17:9. Vaccine. 2004;22:2757-2760.
CHAPTER 25

Pseudorabies
Jane E. Sykes and Sarah D. Cramer

other infected animals, especially wildlife. The virus can survive


Overview of Pseudorabies on fomites and in carcasses for several weeks. In swine, clini-
First Described: 1902 (Hungary, Aujeszky)1 cal signs depend on the age at exposure and virus strain. Sub-
clinical infections and shedding occur commonly. When older
Cause: Suid herpesvirus-1 (Family Herpesviridae, subfamily
pigs do develop signs, they typically develop respiratory disease.
Alphaherpesvirinae, genus Varicellovirus)
Encephalitis occurs in piglets or in adult pigs that are infected
Affected Hosts: Domestic and feral swine, cattle, horses, with virulent strains. Reproductive disease and fetal loss can
sheep, goats, dogs, cats, bears, raccoons, foxes, hedge- occur in pregnant sows. Recovered pigs develop latent infection
hogs, opossums, deer, Florida panther, jackals, coyotes, of neuronal tissues with reactivation and shedding of virus in
nonhuman primates (macaques, marmosets), chickens, semen and nasal, oral, or vaginal secretions following stress.
pigeons, geese, ducks, buzzards, sparrow hawks, rabbits, Disease in dogs is uncommonly reported. However, clusters
guinea pigs, rats, and mice of affected dogs have been described over the past decade, pri-
Geographic Distribution: Worldwide marily in hunting dogs that contact wild swine from parts of
Europe (France, Germany, Belgium) and the southern United
Mode of Transmission: Most often ingestion of uncooked
States (Florida and Oklahoma).2-7 This has followed global
pork by-products or pig carcasses
expansion of wild swine populations. Disease in dogs may be
Major Clinical Signs (Dogs): Variable, but may include fever, more likely to occur in regions that are densely populated with
lethargy, hypersalivation, muscle stiffness, variable facial wild swine, such as the southern United States (Figure 25-1)
pruritus, ataxia, behavioral changes, vomiting, diarrhea, and parts of Europe such as Germany, France, Spain, Poland,
and/or respiratory distress and the Czech Republic.2 Dogs that reside on swine farms may
Differential Diagnoses: Rabies, canine distemper, toxicoses also become infected.8 In some reports, packs of dogs have been
(e.g., organophosphates, heavy metals, ethylene glycol, infected after consumption of uncooked offal from infected pigs
strychnine) or pig carcasses.9,10 Dogs of any age, sex, and breed can develop
disease, but dog-to-dog transmission does not occur. Cats can
Human Health Significance: Humans appear to be resistant be infected after ingestion of pork waste or infected rodents,
to infection with suid herpesvirus-1. but reports of naturally occurring Aujeszky’s disease in cats are
extremely rare.11 Because subclinical infections and nonspecific
signs are common in swine, identification of pseudorabies in
dogs may be the first indication that disease is present in a swine
Etiology and Epidemiology herd or wild swine population.

Pseudorabies (Aujeszky’s disease) is a notifiable disease caused Clinical Features


by the alphaherpesvirus suid herpesvirus-1 (SuHV-1). Swine
are the natural hosts for SuHV-1, but other domestic and wild Signs and Their Pathogenesis
mammals, including ruminants, dogs, cats, raccoons, rabbits, After inhalation or ingestion, SuHV-1 replicates in the orophar-
and rodents, are susceptible to infection and fatal encephalitis. ynx and is taken up by sensory nerve endings. It then travels
The name pseudorabies was applied because the clinical signs by retrograde axonal transport to the sensory nerve ganglia
caused by the virus in rabbits resemble those of rabies. SuHV-1 and then to the central nervous system (CNS), which results
has been eradicated from domestic swine through use of vacci- in ganglioneuritis and encephalitis, especially within the brain-
nation programs in many countries, including Finland, Sweden, stem. Infection can also lead to ganglioneuritis of the cardiac
Norway, the Netherlands, Denmark, Austria, Switzerland, Ger- autonomic plexuses with associated myocardial degeneration,12
many, Hungary, Slovakia, the United Kingdom, Canada, the as well as degeneration of intestinal myenteric ganglia.10 Clini-
United States, and New Zealand. The virus continues to circulate cal signs appear after a short incubation period that typically
among wild boar and feral swine, which act as a reservoir for the ranges from 1 to 6 days, but incubation periods up to 10 days
virus.2 Australia is also free of pseudorabies. Aujeszky’s disease have been reported.9 The duration of signs ranges from 6 to
is a widespread problem in domestic swine herds in Asia, Latin 96 hours.13 The most commonly reported sign is hypersaliva-
America, and some parts of Europe.2 tion. This occurs in most affected dogs, although it may not be
SuHV-1 is shed in the saliva and nasal discharges of swine, present early in the course of illness.10,13,14 Other signs include
and transmission can occur by direct contact with these dis- lethargy, fever, dysphagia, gastrointestinal signs, muscle stiff-
charges or aerosols, or ingestion of infected swine carcasses or ness, ataxia, head-pressing, vestibular signs (circling, head tilt,

257
258 SECTION 1  Viral Diseases

FIGURE 25-1  Geographic distribution of feral swine in the United States. (From
Southeastern Cooperative Wildlife Disease Study, College of Veterinary Medicine, The Uni- FIGURE 25-2  Evidence of facial self-mutilation in a 2-year-old intact male Cata-
versity of Georgia. http://128.192.20.53/nfsms. Last accessed May 2, 2012.) houla hunting dog with pseudorabies. Severe exudative dermatitis is present over the
right eye, external ear canal and muzzle. (Reprinted from Cramer SD, Campbell GA, Njaa
BL, et  al. Pseudorabies virus infection in Oklahoma hunting dogs. J Vet Diagn Invest
2011;23:915-923.)
BOX 25-1
Clinical Signs in 25 Dogs with Pseudorabies from atrioventricular dissociation.12 Although facial pruritus is con-
the United States sidered a classic clinical sign, in one case series, it was present in
only 18% of dogs.14 Death occurs within 96 hours (and usually
Ptyalism: 100% 48 hours) after the onset of clinical signs. Sudden death may
Anorexia: 84% occur, possibly as a result of acute myocarditis.9,14
Ataxia: 76%
Wandering: 64% Physical Examination Findings
Tachypnea: 64% Physical examination findings in dogs with pseudorabies include
Dyspnea: 60% mental obtundation, hypersalivation, dehydration, muscle stiff-
Vocalization: 56% ness, tachypnea or respiratory difficulty, and facial pruritus.
Pruritus: 52% Cutaneous excoriations and edema due to self-trauma may be
Neck stiffness: 48% identified (Figure 25-2). Other neurologic signs include ataxia,
Vomiting: 36% head tilt, circling, hyperesthesia, behavioral abnormalities,
Muscle spasms: 36% anisocoria, mydriasis, nystagmus, slow or absent pupillary light
Aggression: 36% responses, recumbency, and coma. Fever may be present early in
Trismus: 28% the course of infection (up to 108°F or 42°C),13 but terminally,
Dysphagia: 24% hypothermia can occur. A variety of cardiac arrhythmias may
Abnormal pupillary light responses: 20% also be identified.
Seizures: 16%
Mydriasis: 12% Diagnosis
Monroe WE. Clinical signs associated with pseudorabies in dogs. J Am Diagnosis of pseudorabies in dogs and cats is based on a com-
Vet Med Assoc 1989;195:599-602. patible history (i.e., exposure to domestic or wild swine in
regions where the virus is present), suggestive clinical signs,
and histopathology and/or virus detection methods such as
nystagmus), recumbency, intense pruritus and self-mutilation, immunofluorescent antibody staining, immunohistochemis-
vocalization, behavior changes such as pacing and aggression, try, virus isolation, or PCR assay on tissues collected at nec-
and tachypnea or respiratory distress (Box 25-1). Uncom- ropsy (Table 25-1). Antemortem diagnosis of pseudorabies in
monly, apparent blindness, facial paralysis, ptosis, photopho- dogs and cats using virus detection methods has not yet been
bia, abnormal facial sensation, and lacrimation occur. Pruritus described.
generally occurs on the face but can also occur on the shoulder
and forelimbs.10,13 The pruritus may result from infection of Laboratory Abnormalities
neurons that innervate those regions.15 Gastrointestinal signs Laboratory abnormalities in dogs with pseudorabies are non-
are common and include vomiting and diarrhea, which may be specific. Mild anemia and moderate leukocytosis due to a neu-
hemorrhagic (e.g., hematemesis or melena).6 Seizures and coma trophilia may be present. Affected hunting dogs have also had
may also occur. A variety of cardiac arrhythmias have been eosinophilia, hyperglobulinemia, hypoalbuminemia, and hyper-
reported in experimentally infected dogs, which include ventric- glycemia.7 In these dogs, concurrent gastrointestinal parasitism
ular premature complexes, ventricular tachycardia, sinus arrest, may have explained the eosinophilia, hypoalbuminemia, and
atrial tachycardia, atrial fibrillation, wandering pacemaker, hyperglobulinemia. Analysis of the CSF may reveal increased
second-degree heart block, T- and P-wave abnormalities, and CSF protein concentration and mononuclear pleocytosis.
CHAPTER 25  Pseudorabies 259

TABLE 25-1
Diagnostic Assays Available for Pseudorabies in Dogs and Cats
Assay Specimen Type Target Performance
Histopathology Necropsy specimens, especially Meningoencephalitis with eo- Necropsy diagnosis
brainstem or ganglia sinophilic intranuclear inclusions;
SuHV-1 antigen with IHC, IFA or
DNA with in situ hybridization
PCR CNS, tonsils, possibly ­biopsies SuHV-1 DNA Unknown whether PCR could be
of self-traumatized skin or used for antemortem diagnosis.
tonsillar swabs Not widely available.
Virus isolation CNS, tonsils, occasionally other SuHV-1 Specialized procedure. False negatives
tissues such as skin ­biopsies or ­occur commonly.
lung tissue

CNS, Central nervous system; IFA, immunofluorescent antibody; IHC, immunohistochemistry; SuHV-1, suid herpesvirus-1.

Microbiologic Tests
Serologic Diagnosis
Although serology using a variety of antibody detection meth-
ods (such as ELISA and virus neutralization) is used to identify
exposure in swine herds, it has not been useful for antemor-
tem diagnosis in dogs and cats. This likely reflects the rapid
course of disease and insufficient time before death for antibody
responses to develop.

Virus Isolation
Although not always successful, SuHV-1 can be isolated from
tissues (such as the CNS and tonsils) in pig kidney or Crandell-
Rees feline kidney cell monolayers. Immunofluorescent anti-
body can then be used to confirm the presence of the virus in
cells. The sensitivity of virus isolation as an antemortem test
(e.g., on saliva or oropharyngeal swabs) is not known. Haired
skin from regions of pruritus may also be suitable.
FIGURE 25-3  Histopathology in pseudorabies encephalitis. Trigeminal ganglion of
Molecular Diagnosis Using the Polymerase Chain Reaction the dog in Figure 25-2. Neutrophilic, lymphocytic, and histiocytic infiltrates are present.
Conventional and real-time PCR assays have been described Ganglion cells are admixed with necrotic cellular debris, and neuronophagia is prominent.
Within a single ganglion cell is an equivocal intracytoplasmic inclusion (arrow). Bar = 50 µm.
for detection of SuHV-1 in brain tissue collected at necropsy.7
(From Cramer SD, Campbell GA, Njaa BL, et al. Pseudorabies virus infection in Oklahoma
Whether PCR assays are useful for antemortem diagnosis is not hunting dogs. J Vet Diagn Invest 2011;23:915-923.)
known, but PCR could be attempted on biopsies of pruritic
regions or tonsillar swab specimens.

Pathologic Findings Treatment and Prognosis


Abnormalities seen on gross postmortem examination in dogs
with pseudorabies include facial excoriations, tonsillar enlarge- Other than supportive care, there is no known treatment for
ment, gastrointestinal mucosal hyperemia and ulceration, pseudorabies in dogs and cats. Disease is almost always rapidly
myocardial hemorrhage, and pulmonary edema and conges- progressive and fatal.
tion. On histopathologic examination, perivascular infiltrates
of lymphocytes, neutrophils, and macrophages may be present Immunity and Vaccination
in the brainstem and medulla oblongata, with neuronophagia,
neuronal degeneration and necrosis, focal gliosis, and eosino- Although vaccines are used in swine to reduce losses where the
philic intranuclear inclusion bodies within neurons and astro- disease is enzootic, vaccines are not available for dogs and cats,
cytes (Figure 25-3). Involvement of the spinal cord was reported because of the sporadic incidence of the disease.
in a cat.11 Other changes in dogs include suppurative tonsillar
inflammation with necrosis, myocardial degeneration, fibri- Prevention
noid vasculitis, and pulmonary edema. Immunohistochemistry,
immunofluorescence, or in situ hybridization can be used to Pseudorabies can be prevented by cooking pork by-products
identify virus within neurons.8 fed to dogs and cats, preventing contact between infected swine
260 SECTION 1  Viral Diseases

and dogs in regions where the disease is enzootic, and possibly 6. Buergelt CD, Romero CH, Chrisman CL. Pseudorabies in two
through control of rodents. dogs. Vet Med. 2000;95:439-442.
7. Cramer SD, Campbell GA, Njaa BL, et  al. Pseudorabies virus
infection in Oklahoma hunting dogs. J Vet Diagn Invest.
Public Health Aspects 2011;23:915-923.
8. Quiroga MI, Nieto JM, Sur J, et al. Diagnosis of Aujeszky’s disease
Humans appear to be refractory to infection with suid virus infection in dogs by use of immunohistochemistry and in-situ
herpesvirus-1. hybridization. Zentralbl Veterinarmed A. 1998;45:75-81.
9. Hugoson G, Rockborn G. On the occurrence of pseudorabies in
SUGGESTED READINGS Sweden. II. An outbreak in dogs caused by feeding abattoir offal.
Cramer SD, Campbell GA, Njaa BL, et al. Pseudorabies virus infection Zentralbl Veterinarmed B. 1972;19:641-645.
in Oklahoma hunting dogs. J Vet Diagn Invest. 2011;23:915-923. 10. Gore R, Osborne AD, Darke PG, et al. Aujeszky’s disease in a pack
Muller T, Hahn EC, Tottewitz F, et al. Pseudorabies virus in wild swine: of hounds. Vet Rec. 1977;101:93-95.
a global perspective. Arch Virol. 2011;156:1691-1705. 11. Hara M, Shimizu T, Nemoto S, et al. A natural case of Aujeszky’s
disease in the cat in Japan. J Vet Med Sci. 1991;53:947-949.
REFERENCES 12. Olson GR, Miller LD. Studies on the pathogenesis of heart
1. Aujeszky A. Über eine neue Infektienkrankheit bei Haustieren. lesions in dogs infected with pseudorabies virus. Can J Vet Res.
Zentralbl Bakteriol I Orig. 1902;32:353-357. 1986;50:245-250.
2. Muller T, Hahn EC, Tottewitz F, et al. Pseudorabies virus in wild 13. Monroe WE. Clinical signs associated with pseudorabies in dogs.
swine: a global perspective. Arch Virol. 2011;156:1691-1705. J Am Vet Med Assoc. 1989;195:599-602.
3. Thaller D, Bilek A, Revilla-Fernandez S, et  al. Diagnosis of 14. Hawkins BA, Olson GR. Clinical signs of pseudorabies in the dog
Aujeszky’s disease in a dog in Austria. Wien Tierarztl Monatsschr. and cat: a review of 40 cases. Iowa State Univ Vet. 1985;47:116-119.
2006;93:62-67. 15. Takahashi H, Yoshikawa Y, Kai C, et al. Mechanism of pruritus
4. Cay AB, Letellier C. Isolation of Aujeszky’s disease virus from two and peracute death in mice induced by pseudorabies virus (PRV)
hunting dogs in Belgium after hunting wild boars. Vlaam Dierge- infection. J Vet Med Sci. 1993;55:913-920.
neeskd Tijdschr. 2009;78:194-195.
5. Toma B, Dufour B. Transmission de la maladie d’Aujeszky
des sangliers aux suides domestiques. Epidemiol Sante Anim.
2004;45:115-119.
CHAPTER 26

Viral Papillomatosis
Jane E. Sykes and Jennifer A. Luff

five novel types have been described that are not fully classified
Overview of Papillomatosis in Dogs and Cats (Table 26-1).14-21 Canis familiaris papillomavirus (canine papil-
First Described: The transmissibility of canine papilloma- lomavirus 1) is associated with oral papillomas and rarely with
viruses was described in the late 1800s; electron micro- cutaneous endophytic (inverted) papillomas and invasive cutane-
scopic descriptions occurred in the late 1960s1,2 ous SCCs.10,22,23 Other canine papillomavirus types have been
associated with cutaneous inverted and exophytic papillomas,
Cause: Papillomaviruses (family Papillomaviridae)
cutaneous pigmented plaques, or cutaneous in situ SCCs. Papil-
Affected Hosts: A variety of animal species, but papillomavi- lomavirus DNA has been detected in a few canine oral SCCs.23,24
rus types are relatively species specific However, most oral and cutaneous SCCs in dogs are negative
Geographic Distribution: Worldwide for papillomavirus DNA. The eight canine papillomavirus types
have been allocated to three different genera, Lambda, Tau, and
Mode of Transmission: Cutaneous or mucosal inoculation
Chi.7,17 Chi papillomaviruses have been associated with cutane-
through trauma
ous plaque formation. Cocker spaniels and Kerry blue terriers
Major Clinical Signs: Exophytic or endophytic papillomas, may be predisposed to cutaneous papillomas, and pug dogs are
pigmented plaques, in situ or invasive squamous cell predisposed to pigmented plaques. Breed predispositions to papil-
carcinomas lomavirus infections may reflect the presence of congenital defects
Human Health Significance: Canine and feline papillomavi- in cell-mediated immunity; however, this is not yet proven.
rus types do not appear to infect humans, although some Papillomavirus infections are uncommonly reported in cats
feline papillomavirus types resemble some human papil- when compared with dogs. They have been associated with
lomavirus types plaque-like lesions (hereafter referred to as feline plaques), dys-
plastic skin lesions, multicentric Bowenoid in situ SCCs (BISC),
cutaneous invasive SCCs, and feline sarcoids (also known as
fibropapillomas). The complete viral genome sequence for
Etiology and Epidemiology two feline papillomaviruses types has been described, as well
as several stretches of additional novel feline papillomavirus
Papillomaviruses cause warts, or papillomas, in a variety of ani- sequences. Feline sarcoids have been associated with feline sar-
mal species (Figure 26-1). They are non-enveloped, icosahedral coid-associated papillomavirus (FeSarPV) infection, for which
viruses with a circular double-stranded DNA genome. Papillo- cattle may be a reservoir.25 Sarcoids have been described in cats
maviruses have also been associated with malignant transfor- from North America, New Zealand, the United Kingdom, Swe-
mations within the skin and mucous membranes (Box 26-1).3 In den, and Australia, and affected cats are often young (<5 years),
humans, certain papillomavirus types are more likely than oth- male, outdoor cats from rural environments. Felis domes-
ers to induce malignant transformation. Papillomavirus DNA ticus papillomavirus 1 (FdPV-1) has been detected in feline
has been detected using PCR assays on the skin of dogs and cats viral plaques.26,27 Felis domesticus papillomavirus 2 (FdPV-2)
and in the oral cavity of dogs with no signs of papillomas4-6; it
is not clear whether this represents subclinical infection or just
carriage of papillomaviruses.7 Because of this, the association
of papillomaviruses with disease in dogs and cats, especially
squamous cell carcinomas (SCCs), remains somewhat unclear.
Papillomaviruses are resistant in the environment and survive
detergents and high temperatures. Exposure to papillomaviruses
is widespread in the dog population based on serologic studies.8
Congenital or acquired deficiencies in cell-mediated immunity
may predispose to papilloma formation.9-13
Papillomaviruses are relatively host species specific. The num-
ber of different papillomavirus types identified in dogs and cats
has expanded dramatically in recent years as a result of the use
of molecular techniques. Correlations appear to exist between
papillomavirus types and clinical manifestations and progression
of disease, although host immune status is also important. Papil-
lomaviruses are classified on the basis of the L1 capsid protein FIGURE 26-1  Structure of a papillomavirus. The virus is non-enveloped and has a
gene sequence. At least nine types occur in dogs; an additional icosahedral capsid composed of two different capsid proteins (L1 and L2).

261
262 SECTION 1  Viral Diseases

BOX 26-1 TABLE 26-1


Spectrum of Clinical Manifestations Associated with Spectrum of Papillomavirus Types and Disease
Papillomavirus Infections in Dogs and Cats Associations in Dogs
CPV Type* Genus Clinical Manifestation
Dogs
Oral papillomatosis 1 Lambda Oral papillomas, cutaneous endo-
Cutaneous exophytic papillomas (warts) phytic papillomas, invasive SCCs
Cutaneous endophytic papillomas (inverted warts) 2 Tau Cutaneous endophytic and exophytic
Pigmented cutaneous plaques papillomas (warts)
Cutaneous in situ SCCs*
3 Chi Cutaneous pigmented plaques, in
Invasive cutaneous SCCs
situ and invasive SCCs
Oral SCCs
4, 5 Chi Cutaneous pigmented plaques
Cats 6 Lambda Endophytic papillomas
Feline viral plaques 7 Tau Exophytic papillomas and in situ
Bowenoid in situ carcinomas SCC
Invasive cutaneous SCCs
8 Chi Cutaneous pigmented plaques
Feline sarcoids
9 Cutaneous pigmented plaques
*SCCs, squamous cell carcinomas. The majority of SCCs in dogs and *CPV, Canine papillomavirus. Five additional types associated with
cats have not been associated with papillomavirus infection. cutaneous pigmented plaques exist that have not yet been definitively
classified at the time of writing. (Luff JA, Affolter VK, Yeargan B, et al.
and other papillomaviruses, including papillomaviruses with Detection of six novel papillomavirus sequences in canine pigmented
homology to human papillomaviruses, have been detected in plaques. J Vet Diagn Invest 2012;24[3]:576-580.)
cats with plaques, BISCs, and invasive SCCs.27-31 FdPV-2 is
strongly associated with feline plaque formation.32 The finding papillomavirus infections, accidental integration of the human
of papillomavirus DNA with homology to the DNA of human papillomavirus (HPV) genome can occur into host cell DNA,
papillomaviruses in cats suggests the possibility of transmission although this does not seem to be necessary for oncogenesis.36
of papillomaviruses between humans and cats; however, addi- Oral papillomas most commonly occur in young dogs and
tional studies are needed.30,31Although papillomavirus DNA often regress spontaneously over a 4- to 6-week period.7 They
has been detected in a feline oral SCC,33 papillomaviruses do can also occur as a result of immunosuppression, such as after
not seem to be responsible for most oral SCCs in cats.34 treatment with cyclosporin, less commonly chronic glucocor-
ticoid treatment, or chronic infection with Ehrlichia canis.
Clinical Features Rarely, papillomas in the caudal oropharynx obstruct respi-
ration (Figure 26-3). Cutaneous pigmented plaques, to which
Signs and Their Pathogenesis pugs and miniature schnauzers are predisposed, are generally
Infection with papillomaviruses follows inoculation of the virus not of clinical significance but in some situations can progress
through microabrasions in the skin caused by physical trauma. to SCCs.19,38,39 Cutaneous exophytic and endophytic papillo-
Initial infection and viral amplification occurs within keratino- mas usually occur as solitary masses.7 Exophytic papillomas are
cytes of the stratum basale.35 As basal keratinocytes undergo proliferative nodules, whereas endophytic papillomas are cup-
differentiation, early viral proteins (E proteins) are expressed shaped lesions with a central, keratin-filled pore (Figure 26-4).
and associate with regulators of the cell cycle to stimulate cell Feline plaques are flat, often pigmented lesions that resemble
cycle progression.35 This leads to enhanced proliferation of canine papillomas histologically. Feline sarcoids are tumors that
cells in the stratum spinosum and stratum granulosum, with resemble equine sarcoids. Multicentric BISCs appear as mul-
accumulation of infected cells and papilloma formation (Figure tiple, hyperpigmented plaquelike lesions.
26-2). With the exception of feline sarcoids, which remain as
latent (or nonproductive) infections, late viral products, includ- Physical Examination
ing the virus capsid proteins (L1 and L2), are first expressed
in cells of the stratum spinosum. Papillomaviruses are nonlytic Canine Oral Papillomatosis
viruses, and mature virions are shed with cells that exfoliate Oral papillomas can occur on the lips, mucocutaneous junctions,
from the stratum corneum or the nonkeratinized cells of the tongue, pharynx, eyelids, and infrequently haired skin7 (see Fig-
mucous membranes. Infected cells develop a characteristic cyto- ure 26-3; Figure 26-5). They may be accompanied by halitosis,
pathic effect, which consists of cytoplasmic vacuolization. Tens ptyalism, and oral discomfort. Early lesions are smooth and
of thousands of virus particles are produced by each infected raised, but as they enlarge they may become cauliflower-like.
cell. Malignant transformation occurs as a result of the effect of They may be pigmented or nonpigmented.
certain viral products (also known as oncoproteins) on host cel-
lular signals that are responsible for regulation of the cell cycle Canine Cutaneous Exophytic Papillomas
and/or apoptosis.36 For example, some papillomavirus types Cutaneous exophytic papillomas are wartlike projections that
produce proteins that degrade p53 and retinoblastoma pro- appear anywhere on the body, but especially the lower limbs
tein (RB), which are important tumor suppressors.37 In human and footpads.
CHAPTER 26  Viral Papillomatosis 263

Dead keratinocyte
releasing viral particles
Dead keratinocyte
Keratin
SC
1 Keratohyaline
granule
SG
Nucleus
Koilocyte
Cytoplasmic
vacuolization
SS
Inclusion
2 body
SB

Basal lamina B
4

A 3
FIGURE 26-2  Schematic representation of the events of papillomavirus infection in the epidermis. A, (1) The primary infection occurs in a cell of the stratum basale (SB) after the virus gains
entry via a microabrasion. The virus replicates and produces only early proteins, which leads to increased cellular proliferation. (2) Progeny of the initial infected cells migrate laterally and move
upwards, where they accumulate in the stratum spinosum (SS) and stratum granulosum (SG). (3) Cellular differentiation ultimately occurs, and large numbers of virions are produced in association
with papilloma development. Virions are shed with exfoliating keratinocytes of the stratum corneum (SC). (4) Detail from 3. B, Immunoperoxidase staining of papillomavirus antigen in a cutane-
ous plaque from a dog. (B courtesy of Luff J, University of California, Davis. In MacLachlan NJ, Dubovi EJ, eds. Fenner’s Veterinary Virology, 4 ed. Burlington, MA: Academic Press [Elsevier]; 2011.)

Canine Endophytic Papillomas Diagnosis


Endophytic (or inverted) papillomas are cup-shaped cutaneous
nodules that range in size from 2 mm to 2 cm in diameter with a The history and gross appearance of oral papillomas in dogs
central depression (see Figure 26-4). Several different variations is generally sufficient for diagnosis. For other papillomas and
have been described, but they are rare and poorly understood.7 plaques, the diagnosis can be confirmed with biopsy and histo-
Single or multiple lesions can occur anywhere on the body. They pathology. A full-thickness biopsy that includes adjacent nor-
have also been described in the interdigital space.9 mal skin should be collected. The presence of papillomaviruses
can be identified using PCR assays, immunohistochemistry, in
Canine Pigmented Plaques situ hybridization, or electron microscopy, although these tech-
Pigmented plaques usually range in size from 1 mm to 1 cm, niques are primarily used on a research basis. Caution should be
but in one dog progressed to larger sizes (up to 8 cm). Multiple used when interpreting the results of PCR assays because papil-
dark, hyperkeratotic flat lesions are typically present on the ven- lomaviruses can be detected on normal skin, so results should
trum or medial aspects of the limbs.39 be interpreted in association with clinical signs. Identification of
virus within lesions using immunohistochemistry, in situ hybrid-
Squamous Cell Carcinomas ization, or electron microscopy is more supportive of causality.
SCCs are often ulcerated, nodular masses that may be found
alongside pigmented plaques or cutaneous papillomas when Microbiologic Tests
malignant transformation occurs (Figure 26-6). Molecular Diagnosis Using the Polymerase Chain Reaction
PCR assays can be used to detect papillomaviruses within
Feline Sarcoids biopsy or cytobrush specimens from dogs and cats. Rolling
Feline sarcoids are solitary, firm, ulcerated, or nonulcerated circle amplification has been used on fresh biopsy specimens
nodules that occur most commonly on the nasal philtrum, to amplify whole circular genomic DNA from papillomavirus
nares, upper lip, digits, or tail tip, but have also been reported at lesions.7
other locations such as the eyelids and pinnae (Figure 26-7).25,40
Pathologic Findings
Feline Plaques and BISC Histopathology of canine exophytic papillomas generally
In cats, plaques are usually a few millimeters in diameter or lin- reveals epidermal hyperplasia (acanthosis), extensive hyper-
ear lesions. They are usually pigmented and occur on the head, keratosis, and clumped keratohyalin granules in the stratum
neck, dorsal thorax, or abdomen. They can be ulcerated.41 It spinosum. The term koilocytosis is used to describe cells with
may be difficult to differentiate plaques from BISCs, which also papillomavirus-induced cytopathic effects, which consist of
appear as pigmented plaques with crusting and ulceration. The extensive cytoplasmic vacuolation and nuclear pyknosis. Intra-
two may occur together simultaneously. nuclear inclusion bodies may also be present; these occur in the
264 SECTION 1  Viral Diseases

FIGURE 26-4  Inverted papilloma on the abdomen of a flat-coated retriever.


(From Lange CE, Favrot C. Canine papillomaviruses. Vet Clin North Am Small Anim Pract
2011;41:1183-1195.)

C B
FIGURE 26-3  A, Oral papillomas in the pharyngeal region of a 5-year-old male neu- FIGURE 26-5  A, Adult male neutered Rottweiler with a high antibody titer to
tered Boston terrier that was receiving chronic glucocorticoid treatment for a temporal Ehrlichia canis, malnutrition, gastrointestinal parasitism, and sarcoptic mange. B, Oral
lobe glioma. B and C, Histopathology of the lesions revealed hyperkeratosis with increased papillomatosis was present on oral examination and involved the hard and soft palates,
keratohyalin granules and multifocal intracytoplasmic inclusion bodies, as well as second- buccal mucosa, and lips.
ary bacterial infection (not shown). (Courtesy of the University of California, Davis Veteri-
nary Dermatology and Neurology Services.)
CHAPTER 26  Viral Papillomatosis 265

In situ hybridization techniques can also be used. Crystalline


arrays of virus can also be identified within lesions with electron
microscopy.

Treatment and Prognosis


The outcome of papillomavirus infection depends on the virus
strain and host immunity. Oral and cutaneous papillomas in
young dogs generally regress spontaneously 4 to 8 weeks, and
sometimes 3 months after the onset of clinical signs. However,
they may persist in older dogs, or when there is underlying
immune compromise, and occasionally progress to malig-
nancy. If possible, underlying immunosuppression should be
treated (for example, through removal or tapering of immuno-
suppressive drug treatment). Lesions can be removed surgically
if they cause physical obstruction or for cosmetic reasons, but
FIGURE 26-6  Right thoracic limb digital squamous cell carcinoma in a male neutered surgical treatment may be followed by recurrence. Treatment
Australian shepherd after amputation. A large, 4 cm × 5 cm ulcerated mass is present. with azithromycin (10 mg/kg PO q24h for 10 days) appeared
Immunohistochemistry for papillomavirus was negative, but Canis familiaris papilloma- to be effective in a prospective, randomized, double-blinded
virus 2 was detected with PCR and sequencing. (Courtesy of the University of California, placebo-controlled clinical trial that included 12 dogs with
Davis Veterinary Anatomic Pathology Service.)
oral papillomatosis and 5 dogs with cutaneous papillomato-
sis.42 The use of topical or intralesional immunomodulators
(such as imiquimod cream) and antiviral drugs (such as cidofo-
vir) has shown promise for treatment of persistent papillomas
in humans, but these treatments have not been evaluated in
dogs and cats.
Pigmented plaques tend to persist, but most are not associated
with malignant transformation. Transformation to a well-differ-
entiated SCC was reported in an affected miniature schnauzer.43
Feline viral plaques have the potential to transform to Bowenoid
SCC in situ and invasive SCCs.32 Feline sarcoids often recur
locally after surgical removal, but metastasis does not occur.
As more information becomes available, certain papillomavirus
types may be more firmly linked to the presence or absence of
malignant transformation, which may aid prognostication.

FIGURE 26-7  Sarcoid of the lip in an 8 year-old female spayed domestic longhair Immunity and Vaccination
at necropsy. The cat was euthanized because of oral pain and inability to eat and drink.
(Courtesy of the University of California, Davis Veterinary Anatomic Pathology Service.) Recovery from oral papillomatosis is associated with papilloma-
virus type-specific resistance to reinfection. Immunity to papil-
upper layers of the epidermis (see Figure 26-3, C). Endophytic lomaviruses comprises (1) the host immune response to the virus
papillomas are cup-shaped masses of epithelial hyperplasia that and (2) a cell-mediated immune response to the tumor, which is
occur below the level of the surrounding normal skin. Koilo- ultimately responsible for tumor regression. This explains why
cytosis and intranuclear inclusions are often prominent. His- new tumors may not occur, but old tumors persist and continue
topathology of pigmented plaques are characterized by locally to grow in the face of an immune response to the virus, and
extensive epidermal hyperplasia, hyperkeratosis, hyperpigmen- immunization with the virus does not induce tumor regression.
tation, and clumped keratohyalin granules; koilocytes and viral Papillomaviruses can evade the host immune response because
inclusions are generally not observed.7 Feline sarcoids have der- infection is limited to the skin, and the immunogenic capsid pro-
mal fibroplastic proliferations with overlying epithelial hyper- tein is not expressed until the virus reaches the terminally differ-
plasia that includes long, thin rete ridges that extend into the entiated outer layer. A variety of other mechanisms of immune
tumor; they may be confused with sarcomas.40 Feline plaques evasion have been identified in human papillomavirus infec-
exhibit moderate to marked epidermal hyperplasia and koilo- tions, such as interference with interferons and inhibition of
cytosis. Intranuclear inclusions are sometimes present. In situ chemokines such as Il-8 and Il-18 by papillomavirus proteins.44
SCCs are characterized by epithelial hyperplasia, dysplasia, and Viral persistence is necessary for malignant transformation.
increased numbers of mitotic figures, with an intact basement
membrane. Loss of the basement membrane and invasion of the Prevention
dermis indicates invasive SCC.
With the exception of feline sarcoids, immunohistochemis- No vaccines are available for prevention of papillomatosis in
try using antibodies that detect conserved papillomavirus anti- dogs and cats. Prevention of immunosuppressive conditions,
gens (i.e., group-specific papillomavirus antigens) can confirm such as retrovirus infections in cats, and cautious use of potent
the presence of papillomaviruses within lesions. Canine and immunosuppressive drugs such as cyclosporin can also reduce
feline papillomavirus-specific reagents are not widely available. the chance of papillomatosis.
266 SECTION 1  Viral Diseases

Public Health Aspects of more than one different human papillomavirus type has been
detected in epithelial lesions in cats.30,31 Further work, includ-
More than 100 papillomavirus types infect humans. Papilloma- ing whole viral genome sequencing, is required to determine
viruses are relatively species specific, and there is no evidence whether cats can be a source of some papillomavirus infections
that canine papillomaviruses infect humans. However, the DNA for humans and vice versa.

CASE EXAMPLE Monocytes 758 cells/µL (150-1200 cells/µL)


Eosinophils 1704 cells/µL (0-1500 cells/µL)
Platelets 322,000 platelets/µL (150,000-400,000 platelets/µL)
Signalment: “Oscar,” an adult male neutered Rottweiler mix Rare macroplatelets observed.
(see Figure 26-5) Serum Chemistry Profile:
History: Oscar was brought to the University of California, Sodium 144 mmol/L (145-154 mmol/L)
Davis, Veterinary Medical Teaching Hospital for examination Potassium 4.8 mmol/L (3.6-5.3 mmol/L)
after adoption by a veterinary student as a stray from an Chloride 108 mmol/L (108-118 mmol/L)
Indian reservation in Arizona. The student was concerned Bicarbonate 26 mmol/L (16-26 mmol/L)
about the possibility of malnourishment, a painful mouth, Phosphorus 5.1 mg/dL (3.0-6.2 mg/dL)
and pruritic skin disease. Since his rescue 5 days previously, Calcium 10.1 mg/dL (9.7-11.5 mg/dl)
the student had been feeding him four times daily with BUN 26 mg/dL ( 5-21 mg/dL)
maintenance commercial wet and dry dog foods. His Creatinine 0.7 mg/dL (0.3-1.2 mg/dL)
appetite had been ravenous and tapeworm segments had Glucose 108 mg/dL (64-123 mg/dL)
been observed in his feces. He had been isolated from other Total protein 7.7 g/dL (5.4-7.6 g/dL)
pets in the household. Albumin 2.9 g/dL (3.0-4.4 g/dL)
Physical Examination: Globulin 4.8 g/dL (1.8-3.9 g/dL)
Body Weight: 25.5 kg ALT 48 U/L (19-67 U/L)
General: Bright and alert with normal mentation, nervous. AST 30 U/L (19-42 U/L)
T = 100.8°F (38.2°C), HR = 90 beats/min, RR = 18 breaths/min, ALP 117 U/L (21-170 U/L)
mucous membranes pink, CRT = 1 s. Gamma GT <3 U/L (0-6 U/L)
Integument: A dull haircoat was present. Patchy generalized Cholesterol 210 mg/dL (135-361 mg/dL)
alopecia with severe scaling, crusting, hyperpigmentation, Total bilirubin 0.1 mg/dL (0-0.2 mg/dL).
ulceration, and lichenification was present on the Urinalysis: SGr 1.048; pH 5.0, negative protein (SSA), 1+
face, pinnae and pinnal margins, shoulders, trunk, and bilirubin; no hemoprotein, ketones or glucose; 1-2 WBC/HPF,
extremities. rare RBC/HPF, rare bilirubin crystals, moderate sperm.
Eyes, Ears, Nose, and Throat: Moderate mucoid discharge Cytology of Superficial and Deep Skin Scrapings: Sarcoptes
was present from both eyes. The incisor teeth were mites were seen.
severely worn. Oral examination also revealed pain Imaging Findings: Lateral and dorsoventral thoracic
on opening the mouth and multiple pink to gray radiographs were unremarkable. Abdominal
pedunculated masses that ranged from a few millimeters ultrasonography revealed a small, 1 cm hypoechoic nodule
to over 1 cm in diameter on the buccal mucosa and in the body of the spleen. The remainder of the abdominal
gingiva, lateral aspect of the tongue, and on the hard and structures was within normal limits.
soft palates (see Figure 26-5, B). These were consistent Microbiologic Testing: Lateral flow ELISA assay for
with oral papillomatosis. antibodies to Ehrlichia canis, Anaplasma phagocytophilum,
Musculoskeletal: Body condition score 1/9. Normal gait. No and Borrelia burgdorferi and Dirofilaria immitis antigen (4Dx
other musculoskeletal abnormalities were noted. SNAP, IDEXX Laboratories, ME, USA): Positive for antibodies
Cardiovascular and Respiratory: No clinically significant to E. canis.
abnormalities were detected. Vector-borne disease serology panel (Babesia canis, A. phagocy-
Gastrointestinal and Genitourinary: Abdominal palpation tophilum, E. canis, Rickettsia spp. IFA): Positive for antibodies
was within normal limits. Intact male with soft, symmetrical to E. canis (1:655,360) and Rickettsia spp. (1:40,960).
testes. Soft feces were noted on rectal examination. Fecal flotation: Giardia spp. cysts (<1 per 40× field), Sarcoptes
Lymph Nodes: Mandibular and superficial cervical nodes were mites (1 to 10 per 10× field).
enlarged (approximately 3 cm); the popliteal nodes were Diagnosis: Canine oral papillomatosis; malnutrition; sarcoptic
sized within normal limits. mange; suspected chronic canine monocytic ehrlichiosis;
Laboratory Findings: previous exposure to spotted fever group rickettsias;
CBC: giardiasis; cestodiasis.
HCT 32.3% (40%-55%) Treatment: Oscar was treated with doxycycline (5 mg/kg
MCV 64.3 fL (65-75 fL) PO q12h for 6 weeks) for ehrlichiosis, topical selamectin
MCHC 36.2 g/dL (33-36 g/dL) (every 2 weeks for a total of 6 weeks) for sarcoptic
WBC 10,390 cells/µL (6000-13,000 cells/µL) mange, praziquantel (5 mg/kg, once) for cestodiasis,
Neutrophils 5995 cells/µL (3000-10,500 cells/µL) cephalexin (22 mg/kg q12h for 4 weeks) for secondary
Lymphocytes 1891 cells/µL (1000-4,000 cells/µL) bacterial pyoderma, and fenbendazole (50 mg/kg PO
CHAPTER 26  Viral Papillomatosis 267

q24h for 3 days) for giardiasis. The owner was instructed parvovirus, canine adenovirus, parainfluenza virus, and
to bathe him with an oatmeal shampoo twice weekly. rabies virus vaccines and neutered.
At a recheck appointment 4 weeks later, Oscar weighed Comments: The gross appearance of the oral lesions in this
31.5 kg. The skin lesions had improved, and new hair dog was sufficient to make a diagnosis of oral papillomatosis.
growth was noted. Pruritus was still present. More oral Papillomatosis has been reported in other dogs with chronic
papillomas were present, and the owner described E. canis infection, but the extent to which ehrlichiosis
worsened halitosis. Mandibular lymph nodes remained contributed to papillomatosis in this dog is unknown. The
enlarged. Treatment with cephalexin and doxycycline was immunosuppression associated with malnutrition may also
continued for an additional 3 weeks. Three weeks later, have played a role. Resolution of lesions occurred within
the dog’s bodyweight was 33.4 kg, the oral papillomas 7 weeks after treatment of the immunosuppressive disorders,
and mandibular lymphadenomegaly had resolved, and but papillomatosis can also resolve spontaneously over the
Oscar was vaccinated with canine distemper virus, canine same time period.

SUGGESTED READING 15. Luff JA, Affolter VK, Yeargan B, et al. Detection of six novel papil-
lomavirus sequences within canine pigmented plaques. J Vet Diagn
Lange CE, Favrot C. Canine papillomaviruses. Vet Clin North Am Invest. 2012;24:576-580.
Small Anim Pract. 2011;41:1183-1195. 16. Delius H, Van Ranst MA, Jenson AB, et  al. Canine oral pap-
illomavirus genomic sequence: a unique 1.5-kb intervening
REFERENCES sequence between the E2 and L2 open reading frames. Virology.
1. Watrach AM. The ultrastructure of canine cutaneous papilloma. 1994;204:447-452.
Cancer Res. 1969;29:2079-2084. 17. Lange CE, Tobler K, Ackermann M, et al. Three novel canine pap-
2. Watrach AM, Hanson LE, Meyer RC. Canine papilloma: the struc- illomaviruses support taxonomic clade formation. J Gen Virol.
tural characterization of oral papilloma virus. J Natl Cancer Inst. 2009;90:2615-2621.
1969;43:453-458. 18. Lange CE, Tobler K, Lehner A, et al. A case of a canine pigmented
3. Munday JS, Kiupel M. Papillomavirus-associated cutaneous neo- plaque associated with the presence of a Chi-papillomavirus. Vet
plasia in mammals. Vet Pathol. 2010;47:254-264. Dermatol. 2012;23(1):76-80:e18-e19.
4. Lange CE, Zollinger S, Tobler K, et al. Clinically healthy skin of 19. Tobler K, Favrot C, Nespeca G, et al. Detection of the prototype
dogs is a potential reservoir for canine papillomaviruses. J Clin of a potential novel genus in the family Papillomaviridae in asso-
Microbiol. 2011;49:707-709. ciation with canine epidermodysplasia verruciformis. J Gen Virol.
5. Munday JS, Kiupel M, French AF, et  al. Amplification of papil- 2006;87:3551-3557.
lomaviral DNA sequences from a high proportion of feline cutane- 20. Tobler K, Lange C, Carlotti DN, et al. Detection of a novel pap-
ous in situ and invasive squamous cell carcinomas using a nested illomavirus in pigmented plaques of four pugs. Vet Dermatol.
polymerase chain reaction. Vet Dermatol. 2008;19:259-263. 2008;19:21-25.
6. Munday JS, Witham AI. Frequent detection of papilloma- 21. Yuan H, Ghim S, Newsome J, et  al. An epidermotropic canine
virus DNA in clinically normal skin of cats infected and non- papillomavirus with malignant potential contains an E5 gene and
infected with feline immunodeficiency virus. Vet Dermatol. establishes a unique genus. Virology. 2007;359:28-36.
2010;21:307-310. 22. Bregman CL, Hirth RS, Sundberg JP, et al. Cutaneous neoplasms
7. Lange CE, Favrot C. Canine papillomaviruses. Vet Clin North Am in dogs associated with canine oral papillomavirus vaccine. Vet
Small Anim Pract. 2011;41:1183-1195. Pathol. 1987;24:477-487.
8. Lange CE, Tobler K, Favrot C, et al. Detection of antibodies against 23. Teifke JP, Lohr CV, Shirasawa H. Detection of canine oral papil-
epidermodysplasia verruciformis-associated canine papillomavirus lomavirus-DNA in canine oral squamous cell carcinomas and p53
3 in sera of dogs from Europe and Africa by enzyme-linked immu- overexpressing skin papillomas of the dog using the polymerase
nosorbent assay. Clin Vaccine Immunol. 2009;16:66-72. chain reaction and non-radioactive in situ hybridization. Vet
9. Goldschmidt MH, Kennedy JS, Kennedy DR, et  al. Severe papil- Microbiol. 1998;60:119-130.
lomavirus infection progressing to metastatic squamous cell carci- 24. Zaugg N, Nespeca G, Hauser B, et al. Detection of novel papillo-
noma in bone marrow-transplanted X-linked SCID dogs. J Virol. maviruses in canine mucosal, cutaneous and in situ squamous cell
2006;80:6621-6628. carcinomas. Vet Dermatol. 2005;16:290-298.
10. Sundberg JP, Smith EK, Herron AJ, et  al. Involvement of canine 25. Munday JS, Knight CG. Amplification of feline sarcoid-associated
oral papillomavirus in generalized oral and cutaneous verrucosis in papillomavirus DNA sequences from bovine skin. Vet Dermatol.
a Chinese Shar Pei dog. Vet Pathol. 1994;31:183-187. 2010.
11. Le Net JL, Orth G, Sundberg JP, et al. Multiple pigmented cutane- 26. Terai M, Burk RD. Felis domesticus papillomavirus, isolated from
ous papules associated with a novel canine papillomavirus in an a skin lesion, is related to canine oral papillomavirus and contains
immunosuppressed dog. Vet Pathol. 1997;34:8-14. a 1.3 kb non-coding region between the E2 and L2 open reading
12. Callan MB, Preziosi D, Mauldin E. Multiple papillomavirus-associ- frames. J Gen Virol. 2002;83:2303-2307.
ated epidermal hamartomas and squamous cell carcinomas in situ 27. Munday JS, Willis KA, Kiupel M, et al. Amplification of three dif-
in a dog following chronic treatment with prednisone and cyclo- ferent papillomaviral DNA sequences from a cat with viral plaques.
sporine. Vet Dermatol. 2005;16:338-345. Vet Dermatol. 2008;19:400-404.
13. Favrot C, Olivry T, Werner AH, et al. Evaluation of papillomavi- 28. Nespeca G, Grest P, Rosenkrantz WS, et al. Detection of novel pap-
ruses associated with cyclosporine-induced hyperplastic verrucous illomaviruslike sequences in paraffin-embedded specimens of inva-
lesions in dogs. Am J Vet Res. 2005;66:1764-1769. sive and in situ squamous cell carcinomas from cats. Am J Vet Res.
14. Yuan H, Luff J, Zhou D, et  al. Complete genome sequence of 2006;67:2036-2041.
canine papillomavirus type 9. J Virol. 2012;86:5966.
268 SECTION 1  Viral Diseases

29. Schwittlick U, Bock P, Lapp S, et al. [Feline papillomavirus infec- 37. Munday JS, Aberdein D. Loss of retinoblastoma protein, but not
tion in a cat with Bowen-like disease and cutaneous squamous cell p53, is associated with the presence of papillomaviral DNA in
carcinoma]. Schweiz Arch Tierheilkd. 2011;153:573-577. feline viral plaques, Bowenoid in situ carcinomas, and squamous
30. Munday JS, Kiupel M, French AF, et al. Detection of papillomavi- cell carcinomas. Vet Pathol. 2012;49(3):538-545.
ral sequences in feline Bowenoid in situ carcinoma using consensus 38. Stokking LB, Ehrhart EJ, Lichtensteiger CA, et  al. Pigmented
primers. Vet Dermatol. 2007;18:241-245. epidermal plaques in three dogs. J Am Anim Hosp Assoc.
31. Anis EA, O’Neill SH, Newkirk KM, et  al. Molecular character- 2004;40:411-417.
ization of the L1 gene of papillomaviruses in epithelial lesions 39. Munday JS, O’Connor KI, Smits B. Development of multiple pig-
of cats and comparative analysis with corresponding gene mented viral plaques and squamous cell carcinomas in a dog infected
sequences of human and feline papillomaviruses. Am J Vet Res. by a novel papillomavirus. Vet Dermatol. 2011;22:104-110.
2010;71:1457-1461. 40. Schulman FY, Krafft AE, Janczewski T. Feline cutaneous fibropap-
32. Munday JS, Peters-Kennedy J. Consistent detection of Felis domes- illomas: clinicopathologic findings and association with papillomavirus
ticus papillomavirus 2 DNA sequences within feline viral plaques. infection. Vet Pathol. 2001;38:291-296.
J Vet Diagn Invest. 2010;22:946-949. 41. Wilhelm S, Degorce-Rubiales F, Godson D, et  al. Clinical, his-
33. Munday JS, Howe L, French A, et al. Detection of papillomaviral tological and immunohistochemical study of feline viral plaques and
DNA sequences in a feline oral squamous cell carcinoma. Res Vet Bowenoid in situ carcinomas. Vet Dermatol. 2006;17:424-431.
Sci. 2009;86:359-361. 42. Yagci BB, Ural K, Ocal N, et  al. Azithromycin therapy of papil-
34. Munday JS, Knight CG, French AF. Evaluation of feline oral lomatosis in dogs: a prospective, randomized, double-blinded,
squamous cell carcinomas for p16CDKN2A protein immuno- placebo-controlled clinical trial. Vet Dermatol. 2008;19:194-198.
reactivity and the presence of papillomaviral DNA. Res Vet Sci. 43. Nagata M, Nanko H, Moriyama A, et al. Pigmented plaques asso-
2011;90:280-283. ciated with papillomavirus infection in dogs: is this epidermodys-
35. Doorbar J. The papillomavirus life cycle. J Clin Virol. 2005;32(suppl plasia verruciformis?. Vet Dermatol. 1995;6:179-186.
1):S7-15. 44. Kanodia S, Fahey LM, Kast WM. Mechanisms used by human
36. Moody CA, Laimins LA. Human papillomavirus oncoproteins: papillomaviruses to escape the host immune response. Curr Cancer
pathways to transformation. Nat Rev Cancer. 2010;10:550-560. Drug Targets. 2007;7:79-89.
CHAPTER 27

Vector-borne and Other Viral Encephalitides


Jane E. Sykes

documented in dogs and cats. Arboviruses are maintained in


Overview of Vector-borne Encephalitides vectors and small rodent and/or avian reservoir hosts, and some
in Dogs and Cats of these reservoir hosts can develop illness in association with
First Described: Eastern equine encephalitis, 1831 (horses, infection. Dogs and cats, as well as other domestic animal and
Massachusetts)1; West Nile virus infection, 1937 (human, human hosts, are generally considered “dead end” hosts and
Uganda)2; tick-borne encephalitis virus, 1931 (humans, do not maintain sufficient viremia to infect arthropods (Figure
Austria)3 27-1). Arboviruses replicate within the arthropod vector and
are transmitted to vertebrate hosts when the vector feeds; this
Causes: RNA viruses that belong to the families Togaviridae
is followed by viremia and, in some cases, viral spread to the
(genus Alphavirus), such as eastern equine encephalitis
central nervous system (CNS) and infection of neurons (neuro-
virus; Bunyaviridae, such as La Crosse virus; and Flaviviri-
invasion), with subsequent neuronal necrosis and inflammation.
dae, such as West Nile virus and tick-borne encephalitis
When disease occurs, it has important public health significance,
virus
because the clinical signs can mimic those of rabies. Because
Affected Hosts: A variety of animal species including horses, humans have the potential to be infected with these viruses,
humans, and dogs serologic evidence of infection of dogs and cats can also have
Geographic Distribution: Follows that of the vector host. East- public health significance.
ern equine encephalitis virus is found in the eastern and
southeastern United States, as well as Central and South
Alphavirus Infections (Family Togaviridae)
America; West Nile virus is found in Africa, Asia, Middle Alphaviruses are mosquito-borne viruses that belong to the fam-
East, Europe, and the United States; tick-borne encephali- ily Togaviridae. Members of this group that are of importance
tis virus is found in continental Europe and Asia to human and animal health include Eastern, Western, and
Venezuelan encephalitis viruses, which primarily cause disease
Mode of Transmission: Primarily mosquitos and for tick-borne
in humans and horses in the Americas; and Ross River virus,
encephalitis virus, Ixodes ticks.
which causes disease in humans in Australia and the Pacific
Major Clinical Signs (Dogs): Fever (sometimes >105°F islands. Although there is one possible report of encephalitis in
[40.6°C]), lethargy, neurologic signs, and sometimes sys- a dog due to Venezuelan equine encephalitis virus,4 only eastern
temic signs such as anorexia and diarrhea equine encephalitis virus (EEEV) is a significant cause of disease
Differential Diagnoses: Rabies, distemper, pseudorabies; in dogs. Other alphaviruses cause subclinical seroconversion
bacterial, protozoal, or fungal meningitis; granulomatous when they infect dogs; in some reports, seropositive cats have
meningoencephalitis; hemic neoplasia with central ner- also been identified.5 Dogs have been used as sentinels for the
vous system involvement presence of the virus in epidemiologic serosurveys.6
EEEV is endemic in swampy areas of the southeastern United
Human Health Significance: Humans are susceptible to States,7 where it is maintained in a variety of avian reservoir
disease caused by many arboviruses. Dogs and cats are hosts. It causes disease in humans, horses, dogs, and other
dead-end hosts and do not serve as reservoirs for human domestic animal species. Different viral lineages exist that vary
infection. Blood and tissues of affected dogs and cats in pathogenicity for humans and horses. Human cases occur in
should be handled with caution. the southeastern, eastern, and Great Lakes regions of the United
States, but especially Florida, Georgia, Massachusetts, and New
Jersey (Figure 27-2, A).8 Most affected dogs are from Georgia,
but one affected dog was from New York state.9 Dogs with
Etiology, Epidemiology, and Clinical Features signs of encephalitis have always been young (<7 months of
age). Disease usually occurs in late spring or summer. Clinical
A large number of viruses that are spread by arthropod vec- signs include fever, lethargy, inappetence, and diarrhea, with
tors (sometimes referred to as arboviruses, for arthropod-borne rapid progression (within 1 to 2 days) to recumbency, tachy-
viruses) can infect and cause encephalitis in dogs and cats pnea, miosis, nystagmus, tremors, seizures, and death.7
(Box 27-1 and Table 27-1). These viruses are all enveloped
RNA viruses that are unstable in the environment and readily Bunyavirus Infections
inactivated with routine disinfectants. Disease is sporadic and Important bunyaviruses for animal and human health are the
most infected dogs and cats show no clinical signs of illness. mosquito-borne viruses Rift Valley fever (genus Phlebovirus)
For many arboviruses, only subclinical infections have been and La Crosse (genus Bunyavirus) viruses; the tick-borne virus

269
270 SECTION 1  Viral Diseases

TABLE 27-1
Potential Causes of Vector-borne Viral and Other Viral Encephalitides in Dogs and Cats
Virus Classification Vector Affected Hosts Geographic Distribution
Eastern equine Family Toga- Mosquitos Horses, humans, dogs, Southeastern (especially Georgia and Florida),
encephalitis viridae, genus other domestic farm eastern, and Great Lakes regions of the
virus Alphavirus animal species United States
Venezuelan Family Toga- Mosquitos Horses, humans, ­possibly Southern United States (Florida), Central and
equine en- viridae, genus dogs South America (especially Colombia, Ven-
cephalitis Alphavirus ezuela, Trinidad, Peru, Ecuador, Mexico)
virus
La Crosse virus Family Bunyaviri- Mosquitos Humans, dogs Eastern and Midwestern United States
dae
West Nile virus Family Flaviviridae Mosquitos Horses, humans, birds, North, Central, and South America; Africa, the
dogs Middle East, Europe, Asia, Australia
Tick-borne Family Flaviviridae Ixodes ticks Humans, horses, dogs; Europe, Asia
encephalitis domestic animals are
virus generally infected
­subclinically
Borna disease Family Bornaviri- None known Horses, cattle, cats, dogs Primarily Sweden, Austria, Germany, and
virus dae Japan

La Crosse virus was named after La Crosse in Wisconsin, where


BOX 27-1 it was first identified as a human pathogen, and is found mostly
in the eastern and midwestern United States (see Figure 27-2, B).
General Features of Arbovirus Infections as They Relate The virus is maintained in rodents, especially Eastern gray
to Dogs and Cats squirrels and chipmunks. It is a major cause of human pediatric
encephalitis in North America, although mortality is low.
Vector-borne enveloped RNA viruses Affected dogs have included an adult dog from Florida and
Most often mosquito-borne three puppies from southern Georgia.12,13 Mild fever, lethargy,
Maintained among small wild vertebrate reservoir hosts, ataxia, vestibular signs, and ultimately seizures occurred in the
especially birds and/or rodents adult dog, and sudden death was reported in the puppies.
Geographic distribution follows that of vector host
Seasonal distribution that reflects activity of the vector Flavivirus Infections
host Vector-borne flaviviruses (flavi = “yellow,” Latin) of impor-
May cause disease in humans and other domestic animals, tance to human and animal health are the mosquito-borne
especially horses viruses Japanese encephalitis virus, yellow fever virus, Murray
Infection of dogs and cats is most often subclinical Valley encephalitis virus, St. Louis encephalitis virus, Wessels-
Dogs and cats are “dead end” hosts bron virus, Dengue viruses, and West Nile virus (WNV), as well
Infection more prevalent in dogs than in cats as the tick-borne viruses tick-borne encephalitis (TBE) virus,
Sporadic reports of disease, especially in young animals louping ill virus, and Powassan virus. Antibodies to some of
(<6 months) these viruses have been detected in dogs and to a lesser extent,
Neurologic signs accompanied by fever and sometimes cats in serosurveys,14-18 but serologic cross-reactivity among fla-
other systemic signs (e.g., diarrhea) viviruses can occur. Although uncommon to rare, WNV in the
Histopathology shows neuronal necrosis, gliosis, and United States and TBE virus in Europe have been reported to
perivascular cuffing within the brain, which is predomi- cause disease in dogs on more than one occasion. One dog from
nantly nonsuppurative. Africa with fatal encephalitis was initially thought to be infected
with Wesselsbron disease virus,19 but the isolate was later con-
firmed to be WNV.20

Crimean-Congo hemorrhagic fever virus (genus Nairovirus); West Nile Virus Infection
and members of the genus Hantavirus, which are not transmit- West Nile virus was first isolated in 1937 from a woman in
ted by vectors and cause hemorrhagic febrile and respiratory ill- the West Nile region of Uganda.21 In the 1950s, the virus
ness in humans. Serologic evidence of infection with some these was recognized in Israel as a cause of human neurologic dis-
viruses has been documented in dogs and cats.10,11 La Crosse ease in the elderly,22 and subsequently was identified in other
virus is the only bunyavirus reported to cause disease in dogs parts of Europe and South Africa. In 1999, it appeared in New
under natural circumstances, but this appears to be rare.12,13 York state, possibly by transport of infected humans, birds, or
CHAPTER 27  Vector-borne and Other Viral Encephalitides 271

FIGURE 27-1  West Nile virus transmission cycle and factors that influence arbovirus epidemiology.

A B

Western European Tick-borne Encephalitis


Eastern European Tick-borne Encephalitis
D Overlapping Distribution
FIGURE 27-2  Geographic distribution of some arboviruses that can cause disease in dogs and cats. A, Regions with eastern equine encephalitis virus transmission. B, Regions with
La Crosse virus transmission. C, Regions with West Nile virus transmission. D, Regions with tick-borne encephalitis virus transmission.

mosquitos.23 WNV then spread explosively westward across hemisphere, Europe, the Middle East, India, Africa, and Austra-
the United States, and by 2003, disease was detected across lia belong to WNV lineage 1; lineage 2 strains circulate in South
the United States to California (see Figure 27-2, C). WNV also Africa and Madagascar. Other lineages have also been reported.
spread into Canada, Mexico, the Caribbean, and Central and Transmission of WNV by mosquitos increases in the warmer
South America.23,24 Several different variants of the virus exist, months, with peak activity between July and October. Birds
which differ in virulence. Strains circulating within the Western (especially passerines such as crows, jays, and brown sparrows;
272 SECTION 1  Viral Diseases

transstadial
transmission
bank voles
field mice

larva nymph

cofeeding
transstadial
transmission

eggs
transovarial
Ixodes ricinus
transmission
adult tick

FIGURE 27-3  Transmission cycle for tick-borne encephalitis virus.

shorebirds; owls; and hawks) are important reservoir hosts (see Tick-Borne Encephalitis Viruses
Figure 27-1). Some avian species, such as brown sparrows, TBE virus is a notifiable disease that causes potentially fatal
maintain high viremias in the absence of clinical signs. Other neurologic disease in humans in Europe and Asia, where it is
modes of transmission occur, such as ingestion through preda- the most important tick-borne viral disease of humans.32 The
tion of infected birds or rodents, blood transfusion, needle-stick virus is transmitted transstadially and to a lesser extent trans-
injuries, and transplacental transmission. Disease primar- ovarially within infected ticks, and ticks can transmit the dis-
ily occurs in birds, humans, and horses. When signs occur in ease to each other while co-feeding, independent of systemic
humans they are often mild and self-limiting and include fever, viremia in vertebrate hosts.33 Consumption of raw milk from
headache, myalgia, fatigue, gastrointestinal symptoms, and a infected ruminants is a lesser mode of transmission to humans
transient rash.25 Rarely, hepatitis, pancreatitis, myocarditis, (Figure 27-3).
rhabdomyolysis, orchitis, uveitis, and chorioretinitis can occur. Three subtypes of TBE virus exist, which vary in virulence
Severe, neuroinvasive disease occurs in fewer than 1% of cases, and geographic distribution. These are designated the Euro-
is most common in the elderly and results in high fever, cervical pean, Siberian, and far-eastern subtypes.34 The far-eastern sub-
rigidity, muscle weakness, seizures, and flaccid paralysis. Neu- type occurs in far eastern Russia, Japan, Korea, and China and
rologic signs result from viral invasion of neurons in the brain- is the most virulent subtype, with mortality rates in humans
stem, deep nuclei, and anterior horns within the spinal cord. that may exceed 20%. In much of Europe, Ixodes ricinus
Clinical signs in horses result from polioencephalomyelitis and ticks spread the European subtype (Western European TBE)
include fever and neurologic signs such as ataxia, weakness, and whereas Ixodes persulcatus ticks transmit the other viral sub-
muscle tremors.26 Approximately two thirds of horses recover. types in northeastern Europe and Asia (Eastern European TBE)
Infected wild birds can exhibit neurologic disease but are usu- (Figure 27-2, D).35 Both Siberian and European subtypes may be
ally found dead. Myocarditis and meningoencephalitis are com- found in eastern Europe. The disease was originally detected in
mon necropsy findings in these birds.27 1937 in Austria, but its geographic distribution has expanded rap-
In dogs, there is widespread serologic evidence of infection, idly so that the European subtype now occurs throughout Europe
but disease is rare. Cats can become infected and seroconvert, as far west as France and possibly Belgium.17 In Europe, some
but illness has not been described. Five clinically affected dogs of the highest incidence rates have occurred in central European
have been described. All, apart from the dog from Africa, countries, especially the Czech Republic.36 Disease seasonality cor-
resided in various parts of the United States (Missouri, Mis- relates with the activity of the tick in various parts of Europe. TBE
sissippi, California, and Illinois).28-31 All of the dogs had signs has not been reported in the United Kingdom, but a related tick-
of fever and progressive neurologic disease. Neurologic signs borne flavivirus that mainly causes disease in sheep, Louping ill
included generalized tremors, ataxia, a head tilt, bobbing move- virus, is present. This virus rarely causes encephalitis in dogs.37,38
ments, altered mentation, cervical pain, abnormal placing reac- In humans, signs of TBE occur in fewer than 0.5% of
tions, and tetraparesis. Diarrhea, serous oculonasal discharge, infected individuals 4 to 28 days after a tick bite and are
conjunctivitis, abdominal pain, cardiac arrhythmias due to biphasic. In the first, viremic phase, fever, malaise, headache,
myocarditis, tachypnea, tachycardia, and a stiff gait associated myalgia, and vomiting occur, which resolve within 1 week. In
with mild neutrophilic polyarthritis have been reported. For all some individuals, after an asymptomatic period that lasts 2 to
dogs, diagnosis was made at necropsy. 10 days, clinical signs return and are accompanied by signs of
CHAPTER 27  Vector-borne and Other Viral Encephalitides 273

meningitis, meningoencephalitis, meningoencephalomyelitis, Diagnosis


or meningoencephaloradiculitis. About 1% of cases are fatal,
most often in the elderly,39 but neurologic signs can persist Laboratory Abnormalities
for months or longer than a year. Co-infections with Borrelia Laboratory abnormalities in dogs with vector-borne viral
burgdorferi can occur. Vaccines are currently recommended encephalitis are nonspecific. No or minimal abnormalities may
for prevention of TBE in humans, and national vaccination be detected in some dogs. Mild anemia, leukopenia, lympho-
programs in Austria have greatly reduced the incidence of penia, or leukocytosis due to a neutrophilia (sometimes with
disease.40 a left shift) may occur. The CSF may also be unremarkable, or
Subclinical infection of dogs with TBE virus is widespread, there may be increased protein concentration and mononuclear
and so dogs have been important sentinels for the presence of pleocytosis.
the virus in new regions of Europe. Rarely, illness that resem-
bles that in humans can occur in dogs. Affected dogs have been Microbiologic Tests
described from Sweden, Switzerland, Austria, Germany, and Serologic Diagnosis
Italy.32,41 Several reports have described disease in Rottweilers, Serologic assays such as ELISAs are available for detection of
but it is not clear whether this represents a true breed predis- antibodies to arboviruses through specialized veterinary diag-
position. Signs in dogs include high fever, lethargy, anorexia, nostic or research laboratories, and have primarily been used
and neurologic signs such as decreased mentation, behavioral for epidemiologic serosurveys of dogs and cats. Serologic assays
changes such as aggression, delayed placing reactions, ataxia, have, in general, not been very useful for diagnosis of acute
flaccid paralysis, reduced segmental reflexes, neck pain, vestibu- viral encephalitis in dogs and cats. First, because disease is rare,
lar signs, tremors, tetraparesis, miosis, anisocoria, and general- arbovirus infections are often not suspected before death or
ized seizures.32,35 These signs progress rapidly over several days euthanasia, and euthanasia is often performed early because of
to a week, usually culminating in death or euthanasia. Recovery suspicion for rabies. Second, incubation periods are often short
has been described rarely. and disease in dogs progresses rapidly with a high mortality
rate, so there is insufficient time for an antibody response to
Bornaviridae develop. Serologic assays have mainly been used to detect IgG
Borna disease virus (BDV) is an enveloped RNA virus that causes and IgM in dogs with TBE from Europe.32
meningoencephalitis primarily in horses in Europe, but also in
sheep, cattle, and a variety of other domestic and wild animal Virus Isolation
species. On rare occasions, cats and dogs are affected.42,43 BDV Arboviruses can be isolated in a variety of cell lines, and virus
has been suggested as a possible but unconfirmed cause of neu- isolation has been used to detect WNV, TBE virus, and
ropsychiatric illness in humans.44 The epidemiology of BDV La Crosse virus in naturally infected dogs with encephalitis
remains mysterious. There is some evidence that small mammals and BDV in cats.12,28,47,51 Fresh or fresh-frozen brain or spi-
(rodents) act as reservoir hosts. In one study, cats at risk were nal cord homogenates are suitable specimens. Because viremia
intact male cats, cats that hunted, and cats that resided in a rural occurs, blood, CSF, lung, spleen, and liver might also be useful
habitat.45 BDV can be transmitted horizontally among labora- specimens for isolation.28,32 Immunofluorescent antibody stain-
tory rodents via urine.46 Because of seasonal disease occur- ing and reverse transcriptase–PCR (RT-PCR) assays are used to
rence in the spring and summer, an arthropod vector has been confirm the presence of the virus in cell cultures.7,28,47 Because
suspected, but like rabies virus, BDV is thought to enter the these viruses can infect humans, specialized biosafety level 3
CNS principally via axonal transport from mucosal surfaces, facilities are generally required. This limits the availability and
which suggests that direct transmission may be more important. practicality of virus isolation for routine diagnostic purposes.
Entrance via the olfactory system has been suggested based on
lesions found in horses.44 Molecular Diagnosis Using the Polymerase Chain Reaction
Feline infection was first recognized in Sweden in the early Conventional and real-time RT-PCR assays have been described
1970s. The virus has also been detected in cats with neuro- for many different arboviral pathogens and BDV. Specialized
logic disease from Austria, Switzerland, the United Kingdom, veterinary diagnostic and research laboratories generally per-
and Japan.47-49 Some studies have shown a higher prevalence form these assays. In dogs, RT-PCR assays have been used to
of antibodies to BDV in cats with neurologic signs than in cats detect EEEV, WNV, and BDV in clinical specimens.7,28,42,43
without neurologic signs. Affected dogs have been reported Because BDV can be detected using PCR assays in tissues in the
from Japan and Austria.42,43 Serologic evidence of infection has absence of clinical signs, other methods that localize virus to
been detected in a low percentage (3.7%) of cats that lacked sites of inflammation, such as immunohistochemistry or in situ
neurologic disease from the western United States.50 hybridization, are necessary for diagnosis of BDV encephalitis.
The clinical signs of BDV infection can be explained by
the predilection of the virus for the limbic system, brainstem, Pathologic Findings
and basal ganglia. In cats, the disease has been referred to as In general, histopathology of the CNS in dogs with vector-borne
“staggering disease” and is manifested by behavioral changes viral infections, and cats and dogs with Borna disease, reveals
and increased vocalization, anorexia, lethargy, fever, ataxia, nonsuppurative meningoencephalitis or meningoencephalo-
posterior paresis, inability to retract the claws, hypersaliva- myelitis, with multifocal mild to moderate gliosis, neuronal
tion, and seizures. Clinical signs generally progress over 1 to necrosis, and perivascular cuffing with lymphocytes, histio-
6 weeks, which is followed by death or euthanasia or perma- cytes, and plasma cells within Virchow-Robin spaces.12,32,43
nent residual neurologic dysfunction. Affected dogs have had Neutrophils predominate within the meninges of dogs infected
anorexia, lethargy, tremors, salivation, mydriasis, circling, and with EEEV.7 Within brain tissue, inflammatory infiltrates in
seizures.42,43 dogs infected with EEEV have been most prominent in the gray
274 SECTION 1  Viral Diseases

A A

B
B
FIGURE 27-5  Severe lymphocytic and neutrophilic myocarditis and vasculitis due
FIGURE 27-4  Spinal cord segment of a dog with WNV encephalitis that shows gliosis to WNV infection with focally extensive hemorrhage and myonecrosis. A, H&E stain. B,
and multiple degenerate neurons. A, H&E stain. B, Immunohistochemical stain showing Immunohistochemical stain showing WNV immunoreactivity within neutrophils and his-
WNV immunoreactivity (brown). (From Cannon AB, Luff J, Brault C, et al. Acute encepha- tiocytes (brown). (From Cannon AB, Luff J, Brault C, et al. Acute encephalitis, polyarthri-
litis, polyarthritis and myocarditis associated with West Nile virus infection in a dog. J Vet tis and myocarditis associated with West Nile virus infection in a dog. J Vet Internal Med
Internal Med 2006;20:1219-1223.) 2006;20:1219-1223.)

matter of the cerebral cortices and midbrains. Gray matter has only a handful of infected dogs have been reported, the true
also been preferentially infected in dogs with WNV encepha- prognosis is poorly understood. In virtually all dogs with neu-
litis.28 Pathologic changes in dogs with TBE are most severe rologic signs, diagnosis has been made at necropsy. Some dogs
in the brainstem and the cerebellum; meningitis and spinal with TBE recover with supportive care.32 Glucocorticoids have
cord involvement have also been described.32 BDV preferen- been used to treat some dogs and cats with viral encephalitides,
tially infects the limbic system and brainstem, although the but their use is controversial because of immunosuppression,
cerebral cortex may also be involved, and one dog had diffuse and controlled studies are not available.
disease.39,40,44 Immunohistochemistry has been used to detect
antigen of WNV, La Crosse virus, and TBE virus in CNS and Immunity and Vaccination
other tissues of dogs, and to detect BDV antigen in the CNS
of cats and dogs at necropsy (Figure 27-4).13,28,41-43,47 In situ Exposure to vector-borne viruses appears to confer immunity to
hybridization has been used to detect BDV RNA in the CNS of future infections with the same virus. Vaccines are not available
dogs and cats.43,48 for dogs because disease is uncommon to rare. However, vac-
Extraneural lesions have mainly been reported in dogs cines are available and used widely in endemic areas to prevent
infected with WNV. Lymphocytic and necrotizing myocardi- flavivirus infections in humans (such as TBE, yellow fever, and
tis was present in several dogs (Figure 27-5). Other findings in Japanese encephalitis) and WNV encephalitis in horses.
dogs with WNV infection have included evidence of hepatic and
renal tubular epithelial cell necrosis, neutrophilic pancreatitis, Prevention
and mild plasmacytic synovitis.28-31
The risk of arboviral infections in dogs and cats can be reduced
Treatment and Prognosis through control of mosquito and tick vectors. Keeping pets
indoors, especially at dawn and dusk when mosquitos are
Treatment of dogs with vector-borne viral encephalitis is sup- most active, can also reduce exposure, provided mosquitos
portive only. The prognosis appears to be poor, but because are excluded through the use of proper screens and windows.
CHAPTER 27  Vector-borne and Other Viral Encephalitides 275

For viruses such as WNV and BDV, prevention of predation specifically labeled for cats should be used on cats or on dogs
on dead birds or rodents may also reduce the risk of infec- that are in contact with cats, because of the sensitivity of cats to
tion. Drainage of standing water and removal of containers permethrin toxicosis.53
(tin cans, old tires, flower pots, clogged gutters, bunched tar-
paulins) that act as breeding sites are important for mosquito Public Health Aspects
control. Water in birdbaths should be changed at least weekly.
Weeds and brush should be reduced. Human mosquito repel- Many arboviruses that cause disease in humans also infect dogs
lents that contain DEET are not recommended for pets, because and cats. Dogs and cats are generally not considered reservoirs
they can be toxic.52 Some products labeled for tick prevention for transmission of virus to humans because they do not develop
in dogs and cats may help to repel mosquitos but are not likely high-level or sustained viremia. As noted previously, dogs have
to completely protect against mosquito bites. More information been used as sentinels for the presence of these viruses in epide-
on tick prevention can be found in Chapter 28. Only products miologic studies.

CASE EXAMPLE Serum Chemistry Profile:


Sodium 144 mmol/L (145-154 mmol/L)
Potassium 4.0 mmol/L (3.6-5.3 mmol/L)
Signalment: “Paloma,” a 13-year-old female spayed golden Chloride 110 mmol/L (108-118 mmol/L)
retriever–Irish setter mix from Vacaville in northern Bicarbonate 16 mmol/L (16-26 mmol/L)
California. Phosphorus 3.1 mg/dL (3.0-6.2 mg/dL)
History: Paloma was evaluated at the University of California, Calcium 9.1 mg/dL (9.7-11.5 mg/dl)
Davis, Veterinary Medical Teaching Hospital in September BUN 8 mg/dL (5-21 mg/dL)
for a 3-day history of inappetence and lethargy. Two days Creatinine 0.6 mg/dL (0.3-1.2 mg/dL)
before, physical examination by the referring veterinarian Glucose 86 mg/dL (64-123 mg/dL)
had revealed tachycardia (96 beats/min) and pyrexia (103.1°F Total protein 5.7 g/dL (5.4-7.6 g/dL)
or 39.5°C). Results of routine CBC, biochemistry tests, and a Albumin 2.3 g/dL (3.0-4.4 g/dL)
urinalysis did not reveal clinically significant abnormalities. Globulin 3.4 g/dL (1.8-3.9 g/dL)
The dog was treated with amoxicillin (20 mg/kg PO q12h) ALT 25 U/L (19-67 U/L)
and enrofloxacin (10 mg/kg PO q12h), without improvement. AST 34 U/L (19-42 U/L)
Physical Examination: ALP 53 U/L (21-170 U/L)
Body Weight: 28.6 kg Gamma GT 3 U/L (0-6 U/L)
General: Obtunded, 5% dehydrated. T = 106°F (41.1°C), Cholesterol 236 mg/dL (135-361 mg/dL)
HR = 140 beats/min, RR = 92 breaths/min, mucous Total bilirubin 0.3 mg/dL (0-0.2 mg/dL).
membranes pink, CRT = 1 s. Imaging Findings:
Integument: No clinically significant abnormalities. Thoracic, Abdominal, Spinal, and Joint Radiographs: No
Eyes, Ears, Nose, and Throat: Bilateral enophthalmos, significant abnormalities were noted.
mild serous ocular discharge, conjunctivitis, and nuclear Abdominal Ultrasound: Mild mesenteric and moderate
sclerosis were present. Moderate to severe dental disease sublumbar lymphadenomegaly was identified. The dog
was present. Examination of the ocular fundus showed no exhibited pain on ultrasound of the right cranial abdomen, but
abnormalities. no abnormalities in this region were identified. Impressions:
Musculoskeletal: Body condition score 4/9. Ambulatory on all mesenteric and sublumbar lymphadenomegaly.
four limbs, but generalized weakness was noted. Moderate Microbiologic Testing: Aerobic and anaerobic blood
atrophy of the temporalis, quadriceps, and gastrocnemius cultures (five sequentially collected specimens): negative
muscles was present. Serology for Ehrlichia canis, Anaplasma phagocytophilum, and
Cardiovascular, Respiratory, Gastrointestinal, Urogenital Borrelia burgdorferi antibodies (lateral flow ELISA): negative
and Lymphoid Systems: Tachypnea and tachycardia were Treatment: Paloma was treated with lactated Ringer’s solution
identified. There was mild cranial abdominal discomfort on (3 mL/kg/h IV) with potassium chloride supplementation
palpation. All peripheral lymph nodes were normal sized. (20 mEq/L), amoxicillin (22 mg/kg PO q8h), enrofloxacin
Neurologic Examination: Decreased placing reactions were (10 mg/kg PO q24h), and doxycycline (5 mg/kg PO q12h).
noted in all four limbs, and there was pain on manipulation Physical examination on day 2 revealed persistent pyrexia
of the neck. (105.6°F), tachycardia (108 beats/min), and a stiff gait, but
Laboratory Findings: there was no evidence of joint pain or swelling on palpation.
CBC: Cytologic analysis of synovial fluid collected from both
HCT 37.5% (40%-55%) carpal and tibiotarsal joints using arthrocentesis revealed
MCV 67.3 fL (65-75 fL) mild suppurative and mononuclear inflammation.
MCHC 34.4 g/dL (33-36 g/dL) On day 3, the dog became tetraparetic and was unable to stand
WBC 6050 cells/µL (6000-13,000 cells/µL) for longer than a few seconds. Pyrexia (104.5°F) persisted,
Neutrophils 5627 cells/µL (3000-10,500 cells/µL) and treatment with dexamethasone sodium phosphate (0.2
Lymphocytes 363 cells/µL (1000-4000 cells/µL) mg/kg IV once) and hydromorphone (0.05 mg/kg IV once)
Monocytes 61 cells/µL (150-1200 cells/µL) was initiated. Neurologic examination disclosed mental
Adequate platelet numbers (clumped on smear). dullness, decreased placing reactions in all four limbs, and
276 SECTION 1  Viral Diseases

normal segmental reflexes. Further evaluation for neuro- The cervical spinal cord was the most severely affected. Mild
logic disease, such as imaging of the brain, CSF analysis, or perivascular plasmacytic synovitis was also present. Aerobic
both, was offered, but declined by the owner because of and anaerobic cultures of the liver, spleen, spleen, lung, and
financial limitations. On day 4, the dog was normothermic, synovial fluid were negative. West Nile virus antigen was
and sternally recumbent. Prednisone (1 mg/kg PO q12h) detected with immunohistochemical stains within foci of
and metronidazole (10 mg/kg PO q8h) were added to the encephalomyelitis and myocarditis. The virus was isolated
treatment regime. Despite this treatment, by day 5, Paloma from fresh frozen lung tissue, and WNV RNA was detected
was laterally recumbent and obtunded. The owner elected using RT-PCR in the synovium, synovial fluid, spinal cord,
euthanasia. heart, and kidney tissue.25
Necropsy Findings: Histopathologic findings at necropsy Diagnosis: Encephalitis, myocarditis, and polyarthritis
consisted of severe acute multifocal necrotizing perivascular associated with WNV infection
and random neutrophilic pancreatitis; severe lymphocytic Comments: This was a rare case of encephalitis and
and neutrophilic myocarditis and vasculitis with myonecrosis; myocarditis associated with WNV infection in a dog. Disease
and mild, multifocal gliosis and perivascular to random occurred in September, the time of peak activity for virus
lymphoplasmacytic and neutrophilic polioencephalomyelitis. transmission by mosquitos (July to October).

(From Cannon AB, Luff JA, Brault AC, et al. Acute encephalitis, polyarthritis, and myocarditis associated with West Nile virus infection in a dog. J
Vet Intern Med 2006;20:1219-1223.)

SUGGESTED READINGS 13. Tatum LM, Pacy JM, Frazier KS, et  al. Canine La Crosse viral
meningoencephalomyelitis with possible public health implica-
Cannon AB, Luff JA, Brault AC, et al. Acute encephalitis, polyarthritis, tions. J Vet Diagn Invest. 1999;11:184-188.
and myocarditis associated with West Nile virus infection in a dog. 14. Chang HC, Takashima I, Arikawa J, et al. Biotin-labeled protein-A
J Vet Intern Med. 2006;20:1219-1223. enzyme-linked immunosorbent assay for the detection of Japanese
Farrar MD, Miller DL, Baldwin CA, et al. Eastern equine encephalitis in encephalitis antibody in sera from humans, swine and several ani-
dogs. J Vet Diagn Invest. 2005;17:614-617. mal species. J Virol Methods. 1984;9:143-151.
Murray KO, Mertens E, Despres P. West Nile virus and its emergence in 15. Kokernot RH, Hayes J, Will RL, et  al. Arbovirus studies in the
the United States of America. Vet Res. 2010;41:67. Ohio-Mississippi Basin, 1964-1967. II. St. Louis encephalitis virus.
Pfeffer M, Dobler G. Tick-borne encephalitis virus in dogs—is this an Am J Trop Med Hyg. 1969;18:750-761.
issue? Parasit Vectors. 2011;4:59. 16. Resnick MP, Grunenwald P, Blackmar D, et  al. Juvenile dogs as
potential sentinels for West Nile virus surveillance. Zoonoses Pub-
REFERENCES lic Health. 2008;55:443-447.
1. Hanson RP. An epizootic of equine encephalomyelitis that occurred 17. Roelandt S, Heyman P, De Filette M, et al. Tick-borne encephalitis
in Massachusetts in 1831. Am J Trop Med Hyg. 1957;6(5):858-862. virus seropositive dog detected in Belgium: screening of the canine
2. Smithburn KC, Hughes TP, Burke AW, et al. A neurotropic virus population as sentinels for public health. Vector Borne Zoonotic
isolated from the blood of a native of Uganda. Am J Trop Med Dis. 2011;11:1371-1376.
Hyg. 1940;20:471-472. 18. Lindhe KE, Meldgaard DS, Jensen PM, et  al. Prevalence of tick-
3. Schneider H. Über epidemische Meningitis serosa. Wien Klin borne encephalitis virus antibodies in dogs from Denmark. Acta
Wochenschr. 1931;44:350. Vet Scand. 2009;51:56.
4. Habluetzel JE, Grimes JE, Pigott Jr MB. Serologic evidence of natu- 19. Simpson VR, Kuebart G, Barnard B. A fatal case of Wesselsbron
rally occurring Venezuelan equine encephalomyelitis virus infection disease in a dog. Vet Rec. 1979;105:329.
in a dog. J Am Vet Med Assoc. 1973;162:461-462. 20. Burt FJ, Grobbelaar AA, Leman PA, et  al. Phylogenetic relation-
5. Kay BH, Boyd AM, Ryan PA, et al. Mosquito feeding patterns and ships of southern African West Nile virus isolates. Emerg Infect
natural infection of vertebrates with Ross River and Barmah Forest Dis. 2002;8:820-826.
viruses in Brisbane, Australia. Am J Trop Med Hyg. 2007;76:417-423. 21. Smithburn KC, Hughs TP, Burke AW, et  al. A neurotropic virus
6. Coffey LL, Crawford C, Dee J, et al. Serologic evidence of wide- isolated from the blood of a native of Uganda. Am J Trop Med
spread Everglades virus activity in dogs, Florida. Emerg Infect Dis. Hyg. 1940;20:471-492.
2006;12:1873-1879. 22. Bernkopf H, Levine S, Nerson R. Isolation of West Nile virus in
7. Farrar MD, Miller DL, Baldwin CA, et al. Eastern equine encepha- Israel. J Infect Dis. 1953;93:207-218.
litis in dogs. J Vet Diagn Invest. 2005;17:614-617. 23. Murray KO, Mertens E, Despres P. West Nile virus and its emer-
8. Eastern Equine Encephalitis. 2011. http://www.cdc.gov/eastern gence in the United States of America. Vet Res. 2010;41:67.
equineencephalitis/tech/epi.html.Last accessed December 7, 2011. 24. Morshed M, Tang P, Petric M, et al. West Nile virus finally debuts
9. Cornell University College of Veterinary Medicine EEE found in in British Columbia 10 years after its introduction to North Amer-
dog first case in New York state. 2011. http://ahdc.vet.cornell.edu/ ica. Vector Borne Zoonotic Dis. 2011;11:1221-1224.
news/eee.cfm. Last accessed January 29, 2013. 25. Hayes EB, Gubler DJ. West Nile virus: epidemiology and clinical
10. Dobly A, Cochez C, Goossens E, et al. Sero-epidemiological study features of an emerging epidemic in the United States. Annu Rev
of the presence of hantaviruses in domestic dogs and cats from Bel- Med. 2006;57:181-194.
gium. Res Vet Sci. 2012;92(2):221-224. 26. Schuler LA, Khaitsa ML, Dyer NW, et  al. Evaluation of an out-
11. Shepherd AJ, Swanepoel R, Shepherd SP, et  al. Antibody to break of West Nile virus infection in horses: 569 cases (2002). J Am
Crimean-Congo hemorrhagic fever virus in wild mammals from Vet Med Assoc. 2004;225:1084-1089.
southern Africa. Am J Trop Med Hyg. 1987;36:133-142. 27. Steele KE, Linn MJ, Schoepp RJ, et  al. Pathology of fatal
12. Black SS, Harrison LR, Pursell AR, et al. Necrotizing panencepha- West Nile virus infections in native and exotic birds during
litis in puppies infected with La Crosse virus. J Vet Diagn Invest. the 1999 outbreak in New York City, New York. Vet Pathol.
1994;6:250-254. 2000;37:208-224.
CHAPTER 27  Vector-borne and Other Viral Encephalitides 277

28. Cannon AB, Luff JA, Brault AC, et al. Acute encephalitis, polyar- 41. Weissenbock H, Suchy A, Holzmann H. Tick-borne encephalitis in
thritis, and myocarditis associated with West Nile virus infection in dogs: neuropathological findings and distribution of antigen. Acta
a dog. J Vet Intern Med. 2006;20:1219-1223. Neuropathol. 1998;95:361-366.
29. Buckweitz S, Kleiboeker S, Marioni K, et  al. Serological, reverse 42. Weissenbock H, Nowotny N, Caplazi P, et  al. Borna disease
transcriptase-polymerase chain reaction, and immunohistochemi- in a dog with lethal meningoencephalitis. J Clin Microbiol.
cal detection of West Nile virus in a clinically affected dog. J Vet 1998;36:2127-2130.
Diagn Invest. 2003;15:324-329. 43. Okamoto M, Kagawa Y, Kamitani W, et al. Borna disease in a dog
30. Lichtensteiger CA, Heinz-Taheny K, Osborne TS, et al. West Nile in Japan. J Comp Pathol. 2002;126:312-317.
virus encephalitis and myocarditis in wolf and dog. Emerg Infect 44. Lipkin WI, Briese T, Hornig M. Borna disease virus—Fact and fan-
Dis. 2003;9:1303-1306. tasy. Virus Res. 2011;162:162-172.
31. Read RW, Rodriguez DB, Summers BA. West Nile virus encephali- 45. Berg AL, Reid-Smith R, Larsson M, et  al. Case control study of
tis in a dog. Vet Pathol. 2005;42:219-222. feline Borna disease in Sweden. Vet Rec. 1998;142:715-717.
32. Pfeffer M, Dobler G. Tick-borne encephalitis virus in dogs—is this 46. Sauder C, Staeheli P. Rat model of Borna disease virus
an issue? Parasit Vectors. 2011;4:59. transmission: epidemiological implications. J Virol. 2003;
33. Randolph SE. Transmission of tick-borne pathogens between 77:12886-12890.
co-feeding ticks: Milan Labuda’s enduring paradigm. Ticks Tick 47. Lundgren AL, Zimmermann W, Bode L, et al. Staggering disease
Borne Dis. 2011;2:179-182. in cats: isolation and characterization of the feline Borna disease
34. Ecker M, Allison SL, Meixner T, et  al. Sequence analysis and virus. J Gen Virol. 1995;76(Pt 9):2215-2222.
genetic classification of tick-borne encephalitis viruses from Europe 48. Nakamura Y, Watanabe M, Kamitani W, et al. High prevalence of
and Asia. J Gen Virol. 1999;80(Pt 1):179-185. Borna disease virus in domestic cats with neurological disorders in
35. Leschnik MW, Kirtz GC, Thalhammer JG. Tick-borne encephalitis Japan. Vet Microbiol. 1999;70:153-169.
(TBE) in dogs. Int J Med Microbiol. 2002;291(suppl 33):66-69. 49. Reeves NA, Helps CR, Gunn-Moore DA, et  al. Natural Borna
36. Mansfield KL, Johnson N, Phipps LP, et  al. Tick-borne enceph- disease virus infection in cats in the United Kingdom. Vet Rec.
alitis virus—a review of an emerging zoonosis. J Gen Virol. 1998;143:523-526.
2009;90:1781-1794. 50. Abamaku M, Yee JL, Sykes JE, et al. Borna virus-specific antibod-
37. MacKenzie CP. Recovery of a dog from louping-ill. J Small Anim ies in sera from domestic cats in California. 2012:Submitted for
Pract. 1982;23:233-236. publication.
38. MacKenzie CP, Lewis ND, Smith ST, et al. Louping-ill in a working 51. Gresikova M, Sekeyova M, Weidnerova K, et al. Isolation of tick-
collie. Vet Rec. 1973;92:354-356. borne encephalitis virus from the brain of a sick dog in Switzerland.
39. Vaughn DW, Barrett A, Solomon T. Flaviviruses. In: Mandell GL, Acta Virol. 1972;16:88.
Bennett JE, Dolin R, eds. Principles and Practice of Infectious Dis- 52. Dorman DC. Diethyltoluamide (DEET) insect repellent toxicosis.
eases. 7th ed. Philadelphia, PA: Churchill Livingstone, Elsevier; Vet Clin North Am Small Anim Pract. 1990;20:387-391.
2010:2133-2156. 53. Malik R, Ward MP, Seavers A, et al. Permethrin spot-on intoxica-
40. Kunz C, Heinz FX. Tick-borne encephalitis. Vaccine. 2003;21(suppl 1): tion of cats Literature review and survey of veterinary practitioners
S1-2. in Australia. J Feline Med Surg. 2010;12:5-14.
SECTION 2
Bacterial Diseases

CHAPTER 28

Ehrlichiosis
Jane E. Sykes

Overview of Ehrlichiosis Human Health Significance: All four ehrlichial species can
First Described: Ehrlichia canis was first described in 1935 infect and cause disease in humans. The most important
(Algeria).1 Ehrlichia ewingii was described in 1992 (United human pathogen is E. chaffeensis, but the E. muris-like
States).2 Ehrlichia chaffeensis was described in 1991 agent may also be important.
(United States).3
Cause: E. canis (canine monocytic ehrlichiosis), E. ­ewingii

T
(canine granulocytic ehrlichiosis), and E. chaffeensis he ehrlichioses are a group of tick-transmitted diseases
(human monocytic ehrlichiosis); an E. muris-like organism caused by intracellular, gram-negative bacteria that include
also may infect dogs in the United States.4 Ehrlichia canis, Ehrlichia ewingii, and Ehrlichia chaffeen-
sis. An organism related to Ehrlichia ruminantium, the cause of
Affected Hosts: E. canis causes disease in dogs. E. canis or a heartwater disease in cattle, has also been detected in ill dogs from
closely related organism may cause disease in cats and South Africa,5 and an organism that resembles E. muris has been
humans. E. ewingii causes disease in dogs, humans, and detected in an ill dog and humans from the upper Midwest of the
goats. E. chaffeensis causes disease in humans, possibly United States.4 These organisms form morulae (Latin for “mul-
dogs and goats. The E. muris-like agent causes disease in berry”), a cluster of bacteria, within phagosomes of circulating
humans and possibly dogs. leukocytes. Ehrlichia canis infects monocytes and causes canine
Geographic Distribution: E. canis is present worldwide, but monocytic ehrlichiosis (CME), one of the most important infec-
especially in tropical and subtropical regions. E. ewingii tious diseases of domestic dogs that are exposed to ticks world-
is primarily found in the south-central and southeastern wide. Ehrlichia ewingii is an unculturable bacterium that infects
United States. Most reports of E. chaffeensis infection granulocytes and causes canine granulocytic ehrlichiosis. Ehrlichia
are from the southern and south-central United States. chaffeensis causes human monocytic ehrlichiosis; dogs are a pro-
The E. muris-like agent has been found in the upper posed reservoir for this organism. The geographic distribution of
Midwest. each pathogen is generally restricted to that of their vectors and
Mode of Transmission: Rhipicephalus sanguineus ticks (E. canis), mammalian reservoir hosts.
Amblyomma americanum ticks (E. ewingii and E. chaffeensis). Organisms from the genus Ehrlichia are grouped within
the family Anaplasmataceae. Also within this family are the
Major Clinical Signs: The major clinical signs of E. canis infec- bacteria Anaplasma platys and Anaplasma phagocytophilum,
tion are fever, lethargy, inappetence, weight loss, mucosal which cause canine thrombocytic and granulocytic anaplasmo-
hemorrhages, uveitis, pallor, edema, and sometimes neu- sis, respectively (see Chapter 29); and organisms belonging to
rologic signs. E. ewingii primarily causes fever, lethargy, the genera Neorickettsia (see Chapter 31). Rickettsia rickettsii,
inappetence, and signs of polyarthritis. the cause of Rocky Mountain spotted fever (RMSF), and other
Differential Diagnoses: Other tick-borne diseases (such as spotted fever group rickettsiae belong to a separate family,
granulocytic anaplasmosis, Lyme borreliosis and babe- the Rickettsiaceae (Chapter 30). The families Rickettsiaceae
siosis), ­bartonellosis, leptospirosis, lymphoma, multiple and Anaplasmataceae are phylogenetically related through
myeloma, systemic primary immune-mediated disease the order Rickettsiales (Table 28-1). The recent availability
of complete genome sequences for these organisms has helped

278
CHAPTER 28  Ehrlichiosis 279

TABLE 28-1
Members of the Order Rickettsiales of Clinical Importance in Dogs and Cats

Family Anaplasmataceae Rickettsiaceae


Genus Ehrlichia Anaplasma Neorickettsia Rickettsia
Species E. canis A. phagocytophilum N. helminthoeca R. rickettsii
E. chaffeensis A. platys N. risticii
E. ewingii

United States of America United States of America

Mexico Mexico
Brown Dog Tick Lone Star Tick
(Rhipicephalus sanguineus) (Amblyomma americanum)
Belize Belize
Guatemala Honduras Guatemala Honduras
El Salvador Nicaragua El Salvador Nicaragua
Costa Rica Costa Rica
A Panama B Panama
FIGURE 28-1  A, Distribution of Rhipicephalus sanguineus, which transmits Ehrlichia canis, in the United States, Mexico and Central America. B, Distribution of Amblyomma america-
num, which transmits Ehrlichia ewingii and Ehrlichia chaffeensis. The distribution of E. ewingii infections in dogs more closely matches that of A. americanum than the distribution of E. canis
matches that of R. sanguineus (highest prevalence in the southern states). This is because of chronic E. canis infections that occur in dogs with travel histories to southern states, where the
climate is warmer and R. sanguineus is more prevalent.

to elucidate mechanisms of pathogenesis and host-pathogen Eggs


No transovarial
interactions.5-7 transmission
The severity of clinical signs in animals with ehrlichial infec-
tions depends on factors such as the size of the inoculum, host
immunity, and organism species and strain. Because of shared
arthropod vectors and/or concurrent exposure to multiple vec-
tor ticks, co-infections with more than one rickettsial pathogen, Adult tick
as well as other arthropod-borne pathogens such as Babesia
spp., and Bartonella spp., occur commonly in dogs and may
Larva
complicate the clinical picture.

Ehrlichia canis Infection


Nymph
Etiology and Epidemiology
E. canis is transmitted primarily by the brown dog tick
(­Rhipicephalus sanguineus), one of the most widely distrib-
uted ticks worldwide. Infection has been reported in dogs from FIGURE 28-2  Life cycle of Ehrlichia canis. The organism is transmitted only transsta-
Asia, Africa, Europe, and the Americas. Australia appears to be dially (from larva to nymph to adult) within the tick. Jackals, foxes, and possibly coyotes
free of E. canis infection although occasionally seroreactivity to also act as reservoir hosts. A morula is shown within the cytoplasm of a monocyte as seen
E. canis has been identified in dogs. The DNA of E. canis has been on a blood smear.
detected in other tick species, which include other ­Rhipicephalus
species,8,9 Ixodes ricinus,10 Haemaphysalis spp. ticks,11 and Der- distribution appears to be moving northward, and CME has been
macentor spp. ticks11; experimental transmission has been accom- reported in dogs that lack travel history as far north as the Nether-
plished with Dermacentor variabilis ticks.12 Different strains of lands.13 E. canis is the most common pathogen detected in ticks in
E. canis exist that may vary in virulence. Although R. sanguineus Israel.8 Because of chronic, subclinical infection, dogs can be trans-
is found throughout the United States, it prefers warm climates, ported to non-endemic regions and subsequently develop disease
and so disease is diagnosed most frequently in dogs living in the years later. Tick larvae or nymphs acquire infection when they
southeastern and southwestern states (Figure 28-1). In Europe, R. feed on infected dogs. Jackals, foxes, and possibly coyotes may
sanguineus is primarily found in Mediterranean regions, but its also act as reservoir hosts. E. canis is transmitted transstadially
280 SECTION 2  Bacterial Diseases

(i.e., from larva to nymph to adult) within the tick (Figure 28-2).14
No clear age or sex predilection for CME exists, but German
shepherds are reportedly more susceptible, and prognosis may be
poorer in this breed. Cross-bred dogs may be less likely to develop
disease.15 Although natural infection of cats with E. canis (or one
or more closely related organisms) has been described in North
and South America,16-18 clinical ehrlichiosis is rarely reported.

Clinical Features
Signs and Their Pathogenesis
The course of CME has been divided into acute, subclinical,
and chronic phases, although in naturally infected dogs, these
phases may not be readily distinguishable. Clinical signs of
acute disease occur 8 to 20 days after infection. The organism
multiplies by binary fission within vacuoles of mononuclear
phagocytes; rupture of infected host cells leads to infection of
new cells. Immune-mediated mechanisms are important in the
­pathogenesis of disease, and the presence of the spleen appears
to ­contribute to disease severity.19 The clinical manifestations FIGURE 28-3  Retinal hemorrhage and detachment in a 4-year old female spayed
vary considerably among dogs, which may reflect factors such as Rhodesian ridgeback mix with canine monocytic ehrlichiosis. The dog also had mod-
E. canis strain variation, host immune response, stage of dis- erate to severe, poorly regenerative, macrocytic hypochromic anemia (16.4% with a
reticulocyte count of 49,000), lymphopenia (94 cells/µL), thrombocytopenia (26,000
ease, and concurrent infections. Lethargy, inappetence, fever,
platelets/µL), and large numbers of circulating nucleated red blood cells (75/100 WBC).
and weight loss are most common. Replication of the organ- Systolic blood pressure was within normal limits. (Courtesy of University of California,
ism in reticuloendothelial tissues is associated with general- Davis Veterinary Ophthalmology Service.)
ized lymphadenopathy and splenomegaly. Ocular and nasal
discharges, peripheral edema, and, less commonly, mucosal
and cutaneous petechial and ecchymotic hemorrhages can also In reports of feline ehrlichiosis, clinical and laboratory find-
occur. Bleeding tendencies result from thrombocytopenia and ings have generally been similar to those in dogs; one cat had
platelet dysfunction,20 which may reflect immune-mediated polyarthritis.16
platelet damage.21 Neurologic signs may result from menin-
geal inflammation or hemorrhage. Dogs can recover spontane- Physical Examination Findings
ously from the acute phase within 2 to 4 weeks, after which Common physical examination findings in dogs with CME are
time they may eliminate the infection or remain subclinically lethargy, fever, peripheral lymphadenopathy, and splenomegaly.
infected. Sequestration of organisms within the spleen may Ocular and nasal discharge, mucosal petechial hemorrhages, epi-
occur, and the organisms may evade the host immune system staxis, peripheral edema, and/or neurologic signs may be evident.
through antigenic variation.5 This subclinical phase may persist Ocular abnormalities include anterior uveitis, hyphema, retinal
for months to years. hemorrhage, retinal detachment, and optic neuritis, with ante-
Chronic CME develops in only some infected dogs. Factors rior uveitis being most common (Figure 28-3).24,29 Neurologic
that influence the development of chronic disease are unclear, but signs include twitching, ataxia, seizures, vestibular signs, hyper-
genetics may play a role. The presence of pancytopenia typifies esthesia, and cranial nerve defects. Dogs with chronic ehrlichio-
the severe chronic form of ehrlichiosis, and results from hypo- sis may have thin body condition or diffuse muscle atrophy and
plasia of all bone marrow cells.22 Clinical signs range in severity mucosal pallor. Findings in cats with ehrlichiosis have been simi-
and include lethargy, inappetence, bleeding tendencies, muco- lar to those in dogs, and include lethargy, splenomegaly, lymph-
sal pallor, fever, weight loss, lymphadenopathy, splenomegaly, adenopathy, petechial hemorrhages, and retinal detachment.16,17
dyspnea, anterior uveitis, retinal hemorrhage and detachment,
polyuria/polydipsia, and edema.15,22-24 Polymyositis occurs in Diagnosis
some dogs, which can be manifested by diffuse muscle wasting
and tetraparesis.25 Secondary opportunistic infections such as Laboratory Abnormalities
viral papillomatosis, protozoal infections, and bacterial urinary Complete Blood Count
tract infections can also develop, although the precise underly- Thrombocytopenia and occasionally mild leukopenia and a
ing mechanism of immunosuppression, and how it relates to nonregenerative anemia occur 1 to 4 weeks after infection with
successful persistence of E. canis, has not yet been elucidated E. canis. Mild thrombocytopenia, with increased mean platelet
(see Figure 26-5).26,27 Marked granular lymphocytosis and bone volume, may persist during the subclinical phase. Classically,
marrow plasmacytosis may occur, sometimes accompanied by a dogs with chronic CME are pancytopenic, but more commonly,
monoclonal gammopathy, which may lead to misdiagnosis of nonregenerative anemia and thrombocytopenia are noted. In
lymphocytic leukemia or multiple myeloma, respectively. This some dogs, regenerative anemia or leukocytosis due to a neutro-
has led to the recommendation that all dogs with well-differ- philia and band neutrophils are present. Lymphopenia occurs
entiated lymphocytosis or otherwise unexplained monoclonal in most affected dogs, but in some dogs, moderate to marked
gammopathy be tested for E. canis infection.28 Protein-losing granular lymphocytosis (up to 17,000/µL) can occur. Normo-
nephropathy may develop as a result of immune-complex blastosis, that can exceed 50 nucleated RBC per 100 WBC, may
glomerulonephritis. be present.
CHAPTER 28  Ehrlichiosis 281

In some dogs, morulae are visualized within circulating


monocytes (Figure 28-2). The finding of morulae within mono-
cytes using cytologic evaluation of blood smears is insensitive,
especially in dogs with chronic infection, and does not distin-
guish between E. canis and E. chaffeensis infection. Use of buffy
coat smears, thin smears of blood collected from the margin of
the pinna, or splenic aspirates increases the sensitivity for detec-
tion of morulae. In one study, after careful searching, morulae
were found in only 2 of 19 dogs with CME.22

Serum Biochemical Tests


Serum chemistry abnormalities in chronic ehrlichiosis include
variable hypoalbuminemia, hyperglobulinemia, and elevated
ALT and ALP activities. Most often the hyperglobulinemia is
due to a polyclonal gammopathy.15 Monoclonal gammopathy
can also develop. Less commonly, increases in serum urea nitro-
gen and creatinine concentrations are present.22

Urinalysis
Transient proteinuria, with urine protein:creatinine ratios that FIGURE 28-4  Bone marrow plasmacytosis in an 8-year-old female spayed Labrador
exceed 20 (reference range, <1) have been reported in dogs with retriever with canine monocytic ehrlichiosis. Serum globulin was 12.1 g/dL (reference
range, 2.3-4.4 g/dL), and serum protein electrophoresis revealed a polyclonal gammopa-
acute CME. This can resolve by 6 weeks after infection.30,31 Dogs
thy. Plasma cells have a clock-faced nucleus and a clear area adjacent to the nucleus; two
with chronic CME may also have evidence of proteinuria. Pyuria, plasma cells are identified with arrows. Mild to moderate megakaryocyte hyperplasia,
hematuria, and cylindruria may also be present. mild erythroid hypoplasia, and mild mature granular lymphocytosis were also present.
Romanowsky stain.
Coagulation Profile
In addition to thrombocytopenia, coagulation abnormalities
in dogs with CME include prolongation of the buccal mucosal Serologic Diagnosis
bleeding time (BMBT), decreased platelet aggregation, and pro- Most often, the diagnosis of CME is made using serology, which
longation of the APTT.32,33 may be performed using indirect immunofluorescent antibody
(IFA) testing, ELISA technology, or Western blotting. Using IFA
Cerebrospinal Fluid Analysis testing, which is considered the gold standard, antibodies can be
Dogs with central nervous system (CNS) involvement may have detected between 7 and 28 days after initial infection. Dogs with
increased CSF protein concentrations and lymphocytic pleocy- acute ehrlichiosis may have false-negative test results if sufficient
tosis.24 Although rarely found, morulae may be detected in cells time has not elapsed for antibody production to occur. PCR
within the CSF.34 assays may be helpful for diagnosis in this situation. A positive
initial serum antibody titer may reflect previous exposure, and
Bone Marrow Analysis not necessarily ehrlichial disease. Retesting should be performed
In dogs with chronic CME, bone marrow findings include hypo- 2 to 3 weeks later to demonstrate seroconversion, and results of
plasia or aplasia of all bone marrow elements, decreased iron serology should be interpreted in light of a dog’s clinical signs
stores and marrow plasmacytosis (Figure 28-4). Bone marrow and the results of testing for other potential causes of the dog’s
mastocytosis has also been described.35 Myelofibrosis does not illness. Dogs with chronic E. canis infection frequently have
typically develop in chronic CME.36 Some dogs have normal or extremely high IFA titers, sometimes greater than 1:600,000,
hypercellular marrows. and these antibodies may persist in the face of treatment, sug-
gesting persistence of the organism.22 Seroconversion does not
Diagnostic Imaging generally occur in dogs with chronic disease, although antibody
Plain Radiography titers may decline in some dogs with treatment. High titers do
Thoracic radiographs in dogs with CME often show no signifi- not correlate with the severity of hyperglobulinemia, disease in
cant abnormalities, but sometimes bronchointerstitial infiltrates general, or duration of illness. Because of variability of report-
are present. This may reflect an underlying interstitial pneumonia. ing between laboratories, there is no standard “cutoff” titer
that is used to separate positive and negative results. Serologic
Sonographic Findings cross-reactivity to other Ehrlichia species occurs, which includes
Findings on abdominal ultrasonography are nonspecific and E. ewingii and especially E. chaffeensis. Cross-reactivity to
include splenomegaly, alterations in splenic echotexture, A. phagocytophilum antigens can occur to a lesser extent. In
enlargement and hypoechogenicity of the abdominal lymph areas where other rickettsial agents are endemic, Western blot-
nodes, and scant peritoneal effusion. Increased renal echo- ting has been used in an attempt to confirm that positive anti-
genicity and decreased corticomedullary definition may occur in body titers on IFA are truly to E. canis antigens. However,
dogs with glomerulonephritis. Western blotting is laborious to perform and interpret and not
routinely available, and when E. canis antigens are the target
Microbiologic Tests used, it may be difficult to distinguish between E. canis and E.
Available diagnostic assays for ehrlichiosis in dogs and cats are chaffeensis infection.37 As a result, Western blotting has pre-
listed in Table 28-2. dominantly been used on a research basis.
282 SECTION 2  Bacterial Diseases

TABLE 28-2
Diagnostic Assays Available for Ehrlichiosis in Dogs and Cats
Assay Specimen Type Target Performance
Cell culture Whole blood Ehrlichia canis; Not widely offered or utilized for routine diagnostic pur-
Ehrlichia ewingii poses. Requires several weeks’ incubation.
cannot be cultured
Morula detection Whole blood, buffy E. canis or Low sensitivity (especially for chronic E. canis infection).
coat smears, body E. ­ewingii E. canis morulae cannot be distinguished from those
fluids, ­tissue morulae of E. chaffeensis, and E. ewingii morulae cannot be
aspirates ­differentiated from those of A. phagocytophilum.
­Morulae may be confused with platelets, ­cytoplasmic
granules, phagocytized nuclear material, and
­lymphoglandular bodies.
IFA serology Serum Antibodies to Acute and convalescent serology is required for diagnosis
E. canis of acute infection, because initial results may be ­negative
in dogs with acute disease, and positive results can reflect
previous exposure rather than active infection. Dogs
with chronic infection generally do not seroconvert.
Cross-reactivity occurs to other ehrlichial species and
­occasionally to Anaplasma spp.
ELISA serology Serum Antibodies to E. Rapid, inexpensive, can be performed as an in-practice test.
canis or E. ewingii Similar limitations as for IFA. Lack of quantitation limits
antigens ability to document seroconversion.
Western Serum Antibodies to Technically difficult; primarily used on a research basis to
­immunoblotting specific E. canis or identify serologic responses to specific ehrlichial species.
E. ewingii antigens May be difficult to distinguish antibody responses to
E. canis and E. ewingii.
PCR Whole blood; spleen, E. canis, E. muris, or Confirms active infection. Sensitivity and specificity may
lymph node or bone E. ewingii DNA vary depending on assay design and specimen type.
marrow aspirates; ­Sensitivity for diagnosis of chronic CME may be low.
buffy coat or tissue Assays that specifically detect E. muris are not widely
specimens available on a commercial basis.

CME, Canine monocytic ehrlichiosis; IFA, immunofluorescent antibody.

A variety of ELISA assays have been developed for detec- incidental finding of E. canis seroreactivity in dogs screened
tion of antibodies to E. canis. A point-of-care lateral-flow using these assays for heartworm antigenemia should prompt
ELISA device for the simultaneous detection of canine heart- performance of a thorough physical examination and basic
worm antigen, antibodies to E. canis or E. ewingii, antibodies laboratory testing (CBC, chemistry panel, and urinalysis) to
to Borrelia burgdorferi, and antibodies to Anaplasma spp. in evaluate for thrombocytopenia, hyperglobulinemia, and pro-
canine serum, plasma, or whole blood has been marketed for teinuria. When sick dogs test positive, quantitative serology
use in companion animals (SNAP 4Dx Plus, IDEXX Labora- should be performed so that a titer can be obtained as a base-
tories, Westbrook, ME), which includes recombinant surface line for acute and convalescent serologic testing or, for dogs
proteins of E. canis and E. ewingii on a single spot. Accord- suspected to have chronic CME, to evaluate for titers of very
ing to the manufacturer, when IFA and Western blotting were high magnitude that might be consistent with a dysregulated
used as the gold standard, the sensitivity and specificity of the immune response to the organism.
E. canis antigen for detection of E. canis antibodies in 104
samples positive for E. canis antibodies and 236 samples nega- Molecular Diagnosis Using the Polymerase Chain Reaction
tive for E. canis antibodies was found to be 96.2% and 100%, Whole-blood PCR assays for E. canis DNA is more sensitive for
respectively. Other point-of-care ELISA assays for detec- early diagnosis of CME than IFA or ELISA in dogs with acute
tion of E. canis antibodies have also been developed. A sili- disease. PCR assays are widely available for routine diagnosis of
con disc-based assay is available in the United States through E. canis infection. Several laboratories offer panels that include
Antech Diagnostic laboratories that detects Dirofilaria immi- PCR assays for a variety of different vector-borne pathogens.
tis antigen and antibodies to E. canis, Anaplasma spp., and The results of these assays should be interpreted in light of a
B. ­burgdorferi (Accuplex 4, Antech Diagnostics, Irvine, CA). dog’s history, clinical signs, and the results of appropriate sero-
The performance of this assay for diagnosis of ehrlichiosis has logic assays; the last should be performed to support the results
not been thoroughly investigated at the time of writing. The of PCR testing. PCR assays for E. canis may be performed on
CHAPTER 28  Ehrlichiosis 283

blood, lymph node aspirates, splenic aspirates, or bone marrow.


Convalescent IFA or ELISA testing is much more sensitive than TABLE 28-3
PCR assays for diagnosis of chronic CME.22,38,39 The sensitivity Antimicrobial Drug Doses Used for Treatment of Ehrlichiosis
of PCR assays for diagnosis of CME when performed on bone
in Dogs and Cats
marrow in dogs with chronic ehrlichiosis can range from 25%
to 68%, depending on the laboratory.22 The use of PCR assays
Interval Minimum
in the absence of serology is currently not suitable for screening
Drug Dose Route (hours) Duration (days)
potential blood donors for infection. PCR assays may be useful
to confirm infection in the first week of illness, when serologic Doxycycline 5 mg/kg PO, IV 12* E. canis 28 days;
assays are often negative. Depending on the assay used, when E. ewingii
positive, PCR can also be used to confirm the Ehrlichia species 14 days
involved. Oxytetracycline 7.5-10 IV 12 Until gastroin-
mg/kg testinal signs
Blood Culture abate, and then
E. canis can be cultured in certain cell lines, such as DH82 change to oral
cells. This is time-consuming and generally performed only on doxycycline as
a research basis. above
Pathologic Findings *Or 10 mg/kg PO q24h.
Gross pathologic findings in CME include widespread pete-
chial and ecchymotic hemorrhages, generalized pallor, edema,
lymphadenopathy, and splenomegaly.40,41 Ascites may also be hypokalemia (<3.7 mmol/L), and prolonged APTT (>18.25 s).46
present. On histopathology, lymphoid and plasma cell hyper- Platelet counts generally improve and normalize by 2 weeks
plasia, lymphoplasmacytic infiltrates, and vasculitis may be following institution of therapy. After treatment, titers can
present in numerous organs, such as the brain, eye, spinal cord, decline and become negative in 6 to 9 months. Some dogs
spleen, liver, kidneys, lymph nodes, bone marrow, and lungs. retain high titers for several years. Treatment for these dogs
Histiocytic infiltrates may be found in lymph nodes. With chro- should be based on resolution of platelet counts and improve-
nicity, the proportion of plasma cells increases. Non-suppu- ment of hyperglobulinemia, although hyperglobulinemia can
rative meningitis and perivascular cuffing within the CNS can resolve over several months after treatment is discontinued.
also occur.40-42 Glomerulonephitis may be evident,40 although The use of PCR assays on splenic aspirates could be considered
dogs with acute CME have had interstitial nephritis and elec- to determine if persistent infection is present in these dogs,
tron microscopic abnormalities that are more consistent with but whether ongoing treatment with doxycycline changes the
minimal-change glomerulonephritis, with fusion of podocyte outcome for these dogs is unknown. Platelet counts should
processes.30 Little information is available in regard to the prev- be reassessed 1 and 3 months after discontinuation of ther-
alence and type(s) of glomerulonephritis that develop in dogs apy, because of the potential for relapse or reinfection. Other
with chronic CME. causes of illness (especially other vector-borne diseases such as
babesiosis or bartonellosis) should also be considered in dogs
Treatment and Prognosis that fail to respond to treatment.
Other drugs used to treat CME with variable success are
Antimicrobial Treatment chloramphenicol, imidocarb dipropionate, and enrofloxa-
The treatment of choice for CME is doxycycline (10 mg/kg PO cin.47-50 Ehrlichia canis appears to have intrinsic gyrase-medi-
q24h) (Table 28-3). It was the consensus of the ACVIM Infec- ated resistance to fluoroquinolones,51 so although their use can
tious Disease Study Group that dogs and cats should be treated be associated with clinical improvement, it is not recommended.
for a minimum of 28 days.28 Mixed results have been obtained The antiprotozoal drug imidocarb dipropionate appeared effi-
in studies that have evaluated the efficacy of doxycycline for cacious for treatment of E. canis infection in some studies but
treatment of E. canis infection. One study suggested that acute not others.48,49
infection can be eliminated after treatment for just 16 days.43 At this time, treatment of seroreactive but otherwise healthy
Another study that used ticks to infect dogs showed a failure dogs that have normal routine bloodwork results is controver-
of doxycycline, when given for 14 days, to eliminate the organ- sial, because it is unknown if treatment changes outcome for
ism from subclinically infected dogs.44 In another study, dogs these dogs and has the potential to lead to antimicrobial resis-
with acute and subclinical infections became negative by PCR tance or adverse effects of drug therapy.
assay on blood after 28 days of doxycycline treatment, but dogs
with chronic infections remained intermittently positive. How- Supportive Care
ever, ticks still became PCR positive after they fed on treated For dogs with CME that are dehydrated or anemic, IV flu-
dogs, regardless of the stage of disease when treatment was ids or blood products may be required. Use of erythropoi-
initiated.45 etin and granulocyte colony-stimulating factor together with
Whether or not persistence of infection occurs, most dogs ­prednisone was associated with treatment success in a dog
with acute disease show clinical improvement within 24 to 48 with severe chronic ehrlichiosis in one case report.52 Des-
hours. Dogs with severe chronic disease may not respond to ther- mopressin acetate (DDAVP) (1 mcg/kg SC q24h) for 3 days
apy, or cytopenias may resolve over a period of several months. appeared to be efficacious to treat bleeding disorders in a few
Strong risk factors for mortality in one study were severe leu- dogs with CME,33 and treatment resulted in reduction of the
kopenia (WBC <930 cells/µL), severe anemia (HCT <11.5%), BMBT and APTT. If thrombocytopenia fails to resolve with
284 SECTION 2  Bacterial Diseases

­ oxycycline administration, a short course (up to a week)


d sanguineus have been reported as a result of indiscriminate use
of therapy with immunosuppressive doses of glucocorticoids of these drugs.58
could be ­considered in addition to ongoing therapy with Low dose doxycycline (6.6 mg/kg q24h PO) has also been
doxycycline. used to prevent infection in dogs residing in kennels in which
E. canis infection is a problem. Resistance to doxycycline
Immunity and Vaccination remains a theoretical concern in this situation.
Because of chronic, subclinical infection, blood donor dogs
A vaccine for E. canis infection is currently not available, should be screened with serology (or serology and PCR assays)
but in one study vaccination of dogs with an attenuated for evidence of E. canis exposure or infection. All seropositive
strain dramatically reduced disease in dogs challenged with dogs should be excluded as donors. Transport of chronically
a virulent strain of E. canis.53 The immune response to E. infected dogs to non-endemic regions has the potential to intro-
canis infection is not well understood. Because E. canis is an duce the infection or new strains of E. canis to these regions if
intracellular pathogen, cell-mediated immune responses are the appropriate tick vectors are present.
required for pathogen elimination. In one study, dogs with
acute infection developed CD8+ lymphocytosis that subsided Public Health Aspects
after several weeks, despite organism persistence.27 Cytokine
studies suggest that the immune response may vary with the E. canis DNA has been detected in some human patients with
strain of E. canis involved.54,55 Genetic factors are also likely clinical signs of human monocytic ehrlichiosis,59 suggesting that
to be important determinants of the immune response and E. canis might be a cause of monocytic ehrlichiosis in people.
outcome of infection. Appropriate precautions should be taken to prevent transmis-
sion when handling engorged ticks as well as blood and tissue
Prevention specimens from infected dogs, and care should be taken to pre-
vent needle-stick injuries.
Avoidance of tick-infested areas and routine inspection of dogs
for ticks after outdoor activities can help to prevent ehrlichio- Ehrlichia ewingii Infection
sis. Early removal of ticks may help to reduce transmission,
because of the 24- to 36-hour delay that occurs between tick Etiology and Epidemiology
attachment and feeding. Clients should be instructed to remove
ticks properly and avoid handling ticks with bare hands or Ehrlichia ewingii is an unculturable bacterium that causes
crushing them, to prevent exposure to infected hemolymph. A granulocytic ehrlichiosis in humans and in dogs. It was first
variety of devices are available to assist tick removal, which are recognized in dogs2 and occurs in North America and more
placed around the area where the mouthparts enter the skin to recently has been detected in dogs from Africa and Brazil.60,61
avoid crushing or squeezing the tick and leaving the mouth- Infection occurs in the south-central and southeastern parts
parts behind. Fine-tipped tweezers can be used to grasp the tick of the United States, which reflects the distribution of the
as close to the skin as possible, followed by steady retraction primary tick vector, Amblyomma americanum. Transmis-
to remove the tick. The bite wound should then be thoroughly sion within the tick is transstadial. More than 40% of dogs
cleaned with a suitable antiseptic solution (such as iodine or from an endemic area in Oklahoma and Arkansas were
chlorhexidine-based antiseptics) or soap and water. Ticks can seropositive.62 In another large study the overall seropreva-
be disposed in alcohol or tested for vector-borne pathogens lence was 14.5% in the central states (Oklahoma, Arkansas,
using PCR assays. Missouri, and Kansas) and 5.9% in the southeast.63 In one
Topical ectoparasiticides with activity against ticks also study, most cases occurred from May through July,64 but in
prevent tick-borne infectious diseases. Examples of canine another, cases occurred throughout the year.65 In the study
ectoparasiticides with activity against ticks include those that from the south-central United States, dogs were more likely
contain amitraz, fipronil, pyrethroids (permethrin, etofenprox, to test positive with a PCR assay in August.62 E. ewingii is
pyrethrin, deltamethrin, flumethrin), and selamectin. In one maintained in white-tailed deer.66 The DNA of E. ewingii has
field study, the use of monthly permethrin effectively prevented also been found in Dermacentor variabilis and R. sanguineus
infection with E. canis by kenneled dogs.56 Products that con- ticks,67 but A. americanum is the only proven vector.
tain pyrethroids and amitraz have the most potent activity
against ticks. Permethrin, deltamethrin, and amitraz cannot be Clinical Features
used in cats, and use of these products should be avoided on
dogs that co-habitate with cats. Flumethrin collars are avail- Signs and Their Pathogenesis
able for use on cats (and on dogs) because unlike the other In contrast to E. canis infection, E. ewingii infection appears
pyrethroids, flumethrin does not require hepatic glucaronida- to cause only acute disease; a chronic phase of disease has not
tion for metabolism.57 Products that contain amitraz, a mono- been described. Like A. phagocytophilum, E. ewingii replicates
amine oxidase inhibitor, should not be used in dogs treated in neutrophils and delays neutrophil apoptosis, which prolongs
with selective serotonin reuptake inhibitors (SSRIs) such as the life span of the host cell.68
fluoxetine, and pet owners that are receiving SSRIs should Dogs with E. ewingii infection may show no signs, or fever,
also use alternative preventatives for their dogs. Preventatives lethargy, anorexia, and neutrophilic polyarthritis may occur.
should be applied consistently at the recommended interval for After experimental infection, signs develop after an incubation
optimum activity. No preventatives completely protect dogs period of 3 to 4 weeks.69-71 Vomiting and diarrhea occur uncom-
or cats from tick attachment, especially where tick infestation monly. Neurologic signs have been described in some naturally
rates are high. Unfortunately, acaricide-resistant strains of R. infected dogs,65 but the possibility of concurrent infections with
CHAPTER 28  Ehrlichiosis 285

other pathogens such as Rickettsia rickettsii was not ruled out Subclinically infected dogs that test positive with whole-blood
in these dogs. E. ewingii can be found in apparently healthy PCR assays may spontaneously clear infection within weeks to
dogs, so dogs may act as a reservoir for infection. months. Treatment may not be required for these dogs.

Physical Examination Findings Prevention


On physical examination, dogs with E. ewingii infection may
have evidence of lethargy, fever, lameness, reluctance to move, Prevention of E. ewingii infection involves avoidance of tick
a stiff gait, joint effusion, and pain on joint palpation. Reported exposure and use of tick preventatives (see previously). Early tick
neurologic signs include anisocoria, tremors, and a head tilt.65 removal also has the potential to reduce transmission. Poten-
tial blood donor dogs could be screened for infection with E.
Diagnosis ewingii–specific ELISA and PCR assays. PCR assays alone (i.e.,
without serology) should not be used to screen blood donors.
Laboratory Abnormalities
Common laboratory findings in dogs with E. ewingii infection Public Health Aspects
are nonregenerative anemia and thrombocytopenia.65,72 Reac-
tive lymphocytosis can also occur.65 The biochemistry panel Human infection with E. ewingii has been rarely described in
may be unremarkable or show mild nonspecific abnormali- endemic regions of the United States.73 Affected people have
ties. Synovial fluid analysis reveals neutrophilic polyarthritis. had headache, fever, and thrombocytopenia, with or without
Morulae may be detected within granulocytes in the periph- leukopenia, and responded to doxycycline treatment. Most
eral blood or synovial fluid, but are indistinguishable from were receiving immunosuppressive drug therapy. Although
those of A. phagocytophilum. Nevertheless, the finding of direct dog-to-human transmission does not occur, blood from
morulae in granulocytes in E. ewingii endemic areas and where affected dogs should be handled with caution.
A. phagocytophilum infection is uncommon or absent is
strongly suggestive of E. ewingii infection. In experimental Ehrlichia chaffeensis Infection
infections, morulae are visible in the peripheral blood of some
dogs before the onset of clinical signs, around 2 to 3 weeks Ehrlichia chaffeensis causes human monocytic ehrlichiosis in
postinfection.71 North America, an emerging disease that is characterized in
human patients by fever, headache, myalgia, thrombocytopenia
Microbiologic Tests and leukopenia, and elevations in hepatic transaminases.74 Gas-
Serologic Diagnosis trointestinal signs, neurologic involvement, and a toxic shock–
Serologic diagnosis of E. ewingii infection is currently limited like syndrome also occur in some infected people. In the United
to a point-of-care ELISA assay that detects the presence of States, it occurs primarily in the south-central, southeastern,
antibodies to a specific peptide of E. ewingii (SNAP 4Dx Plus, and mid-Atlantic states, which reflects the distribution of
IDEXX Laboratories, Westbrook, ME). Because this peptide is A. americanum and the concurrent presence of white-tailed
combined on a spot with a peptide that detects antibodies to deer, which are reservoirs for the organism (as for E. ­ewingii).
E. canis, the antibody response to these pathogens cannot Evidence of E. chaffeensis DNA has also been found in
be distinguished from one another. Cross-reactions may also humans, dogs, other animal species (including cats18), and
occur to other ehrlichial species such as E. chaffeensis. Anti- ticks in Africa, Israel, Central and South America, and Asia.
bodies appear approximately 1 month after infection.71 Dogs The organism is transmitted transstadially within the tick,75
with acute illness may initially test negative with this assay, which feeds aggressively on humans. In naturally infected dogs,
and positive test results may reflect the presence of antibodies E. ­chaffeensis infection has been associated with clinical signs
from previous exposure or subclinical infection. A change from of lymphadenopathy, anterior uveitis, and epistaxis,76 but the
a negative to a positive result over a 2- to 4-week time period clinical implications of this infection for dogs are still unclear.
may help support a diagnosis of E. ewingii infection in endemic Dogs maintain high antibody titers and are PCR positive for
areas. months after infection, which supports a possible role of the dog
as a reservoir.77
Molecular Diagnosis Using the Polymerase Chain Reaction
Whole-blood PCR assays are available for specific diagnosis of Ehrlichia muris-like Infection
E. ewingii infection through some veterinary diagnostic labora-
tories. The sensitivity of these assays may vary between labo- Ehrlichia muris was first described in a wild mouse from Japan
ratories. Currently E. ewingii–specific PCR assays are the only in the mid-1990s78,79 and E. muris DNA was detected in a
means to confirm active infection with E. ewingii (as opposed febrile person in Russia for the first time in 2008.80 In 2009,
to infection with A. phagocytophilum or other Ehrlichia spe- the DNA with strong homology to that of Ehrlichia muris was
cies). In experimental studies, PCR becomes positive as early as detected in four humans from Minnesota and Wisconsin.80
4 days after inoculation.71 These individuals had clinical signs and laboratory abnormali-
ties that resembled those of granulocytic anaplasmosis (see
Treatment and Prognosis Chapter 29), were seronegative to A. phagocytophilum, and
showed variable seroreactivity to E. chaffeensis. Two of the
Antimicrobial Treatment individuals were solid-organ transplant recipients and the other
For E. ewingii infection, treatment with doxycycline results two people were apparently immunocompetent. Morulae were
in rapid (within 24 to 48 hours) clinical improvement. Treat- not seen, but in mice E. muris forms morulae in monocytes.78
ment for 2 to 4 weeks may be sufficient to eliminate infection. In 2012, DNA with homology to E. muris was detected in an
286 SECTION 2  Bacterial Diseases

ill dog from northern Minnesota that was seronegative for E. ticks from northern Wisconsin.81 The extent to which E. muris
canis and seroreactive to A. phagocytophilum,4 although the causes disease in dogs and humans in the United States and
extent to which this organism contributed to the dog’s clinical other countries requires further investigation but it should be
signs was unclear. Because of the geographic distribution of considered as a possible cause of unexplained febrile illness
infection, Ixodes scapularis is the suspected tick vector (see in dogs, whether or not they have evidence of antibodies to
Chapter 29). E. muris DNA has been detected in I. scapularis E. canis.

CASE EXAMPLE Chloride 105 mmol/L (108-118 mmol/L)


Bicarbonate 18 mmol/L (16-26 mmol/L)
Phosphorus 6.1 mg/dL (3.0-6.2 mg/dL)
Signalment: “Ditto” a 13-year old male neutered border collie Calcium 8.6 mg/dL (9.7-11.5 mg/dl)
mix from Dixon, CA BUN 55 mg/dL (5-21 mg/dL)
History: Ditto was brought to the University of California, Davis, Creatinine 1.9 mg/dL (0.3-1.2 mg/dL)
because of a 4-day history of lethargy and inappetence. Glucose 66 mg/dL (64-123 mg/dL)
Ditto had been drinking and there was no vomiting or Total protein 6.1 g/dL (5.4-7.6 g/dL)
diarrhea. There was no history of exposure to ticks, toxins, Albumin 2.2 g/dL (3.0-4.4 g/dL)
or trauma. Ditto was mainly confined to the backyard and Globulin 3.9 g/dL (1.8-3.9 g/dL)
did not have access to standing water. The dog had been ALT 49 U/L (19-67 U/L)
adopted in Guam and brought to the United States several AST 153 U/L (19-42 U/L)
years ago. There was no other significant travel history. ALP 98 U/L (21-170 U/L)
Ditto’s diet consisted of a commercial senior dry dog food. Creatine kinase 341 U/L (51-399 U/L)
Current Medications: Prednisolone (0.45 mg/kg PO q48h) for Gamma GT 4 U/L (0-6 U/L)
allergic skin disease; this had been administered over the Cholesterol 282 mg/dL (135-361 mg/dL)
preceding 10 days. Total bilirubin 0.2 mg/dL (0-0.2 mg/dL)
Physical Examination: Magnesium 2.6 mg/dL (1.5-2.6 mg/dL).
Body Weight: 22.3 kg Urinalysis: SGr 1.018; pH 7.0, protein 75 mg/dL, no bilirubin, no
General: Quiet, reluctant to stand. T = 102.2°F (39.0°C), HR = 152 glucose, hemoprotein 25 erythrocytes/µL, 0-2 WBC/HPF, 0-2
beats/min, panting, mucous membranes pale pink, CRT <2 s. RBC/HPF, rare granular casts
Approximately 5% to 7% dehydrated. Urine Protein: Creatinine Ratio: 19.3 (reference range, <1)
Integument: A sparse hair coat was present; there were no Plasma Antithrombin: 64% (reference range, 80%-120%)
other clinically significant abnormalities. No ectoparasites Imaging Findings:
were noted. Thoracic Radiographs: The right cranial mainstem bronchus
Eyes, Ears, Nose, and Throat: Severe dental calculus was was slightly widened throughout its visible length and
present. bronchiectasis was suspected. The cardiovascular and
Musculoskeletal: Body condition score was 6/9. The dog remaining pulmonary structures were within normal limits
would stand but was reluctant to walk. for the age of the patient.
Cardiovascular and Respiratory Systems: No clinically Abdominal Radiographs: An ill-defined soft tissue structure
significant findings were present. was present that displaced the caudal border of the
Gastrointestinal and Urogenital Systems: Cranial stomach cranially. The cecum was not well visualized.
organomegaly was noted on abdominal palpation. Rectal The liver extended past the costochondral arches with
examination revealed no significant abnormalities. mildly rounded borders, which was interpreted as mild
Lymph Nodes: All peripheral lymph nodes were normal sized. hepatomegaly. The soft tissue structure was thought to
Laboratory Findings: represent an enlarged lymph node, pancreas, or spleen.
CBC: Abdominal Ultrasound: The liver had normal parenchymal
HCT 28.0% (40%-55%) architecture. The gallbladder contained hyperechoic
MCV 72.4 fL (65-75 fL) sludge. The spleen was enlarged with rounded edges and
MCHC 30.4 g/dL (33-36 g/dL) a heterogenous, more hypoechoic parenchyma. Both
Reticulocyte count 7600 cells/µL (7000-65,000 cells/µL) kidneys showed a somewhat thickened cortex with a good
Nucleated RBC 1/100 WBC distinction between cortex and medulla. The left renal pelvis
WBC 19,600 cells/µL (6000-13,000 cells/µL) was mildly dilated, and the papilla was blunted. There were
Neutrophils 15,876 cells/µL (3000-10,500 cells/µL) several cystic lesions within the abdomen that measured up
Lymphocytes 392 cells/µL (1000-4000 cells/µL) to 3.6 cm × 5.4 cm. Septa were visualized within the cystic
Monocytes 1568 cells/µL (150-1200 cells/µL) lesions. The cystic lesions appeared to arise from lymph
Eosinophils 392 cells/µL (0-1500 cells/µL) nodes.
Basophils 392 cells/µL (0-50 cells/µL) Cytologic Findings: Cytology of ultrasound-guided lymph
Platelets 127,000 platelets/µL (150,000-400,000 platelets/µL) node aspirate: There was a moderate amorphous basophilic
MPV 20.5 fL (7-13 fL). proteinaceous background with low numbers of nucleated
Serum Chemistry Profile: cells and erythrocytes. Nucleated cells were composed
Sodium 141 mmol/L (145-154 mmol/L) primarily of a mixed lymphocyte population with lower
Potassium 4.2 mmol/L (3.6-5.3 mmol/L) numbers of macrophages and nondegenerate. neutrophils.
CHAPTER 28  Ehrlichiosis 287

Microbiologic Testing: 4Dx SNAP test serology (IDEXX urine protein:creatinine ratio was 4. At a recheck examination
Laboratories, ME): Positive for antibodies to Ehrlichia canis. 1 week after discharge, the hematocrit was stable, the platelet
Weakly positive for antibodies to Anaplasma species. count was 635,000 platelets/µL, BUN 41 mg/dL, creatinine
Negative for antibodies to Borrelia burgdorferi and antigen 1.3 mg/dL, and albumin 2.2 mg/dL. A kidney biopsy was not
of Dirofilaria immitis. performed. Ditto was subsequently managed for protein-
Vector-borne disease serology (IFA): Positive for antibodies to losing nephropathy with enalapril and a reduced protein
Ehrlichia canis at 1:163,840 and Anaplasma phagocytophilum diet for nearly 2 years, at which time hematocrit was 38.9%,
at 1:2560. Negative for antibody to Rickettsia rickettsii at <1:40. platelet count 561,000 platelets/µL, BUN 27, creatinine
Vector-borne real-time PCR panel (whole blood): Positive for 1.2 mg/dL, albumin 3.2 g/dL, globulin 3.6 g/dL, and the urine
Ehrlichia canis DNA. Negative for Anaplasma phagocytophi- protein:creatinine ratio was 1.6. The spleen had returned to
lum, Anaplasma platys, Bartonella spp., Rickettsia spp., and a sonographically normal appearance, and the cystic lymph
Borrelia burgdorferi DNA. nodes were markedly reduced in size. Additional serology or
Diagnosis: Canine monocytic ehrlichiosis, characterized by follow-up PCR was not performed. He was subsequently lost
thrombocytopenia, abdominal lymphadenopathy, and to follow-up.
protein-losing nephropathy. Comments: In this dog, active infection with E. canis was
Treatment: Ditto was initially treated with IV crystalloids confirmed through the use of whole-blood PCR assay.
(lactated Ringer’s solution supplemented with 20 mEq/L KCl), Where the dog became infected with E. canis was unclear,
famotidine (0.5 mg/kg IV q12h), and ampicillin (20 mg/kg IV but infection was most likely acquired many years earlier
q8h). The hematocrit dropped to 18%, and 1 unit of packed in Guam, where ticks are abundant and R. sanguineus and
RBC was administered, which was followed by a transfusion E. canis are present. It is also unclear whether the glucocorticoid
reaction, characterized by hemoglobinuria, vomiting, and treatment played any role in reactivation of chronic
icterus. Systolic blood pressure remained within normal limits infection. The improvement in hematologic and biochemical
throughout hospitalization. When the results of serology parameters in association with doxycycline treatment
for E. canis were obtained, antimicrobial drug treatment supported a role for E. canis in the disease. Hyperglobulinemia
was changed to doxycycline (5 mg/kg PO q12h). Enalapril may have been initially masked by renal protein loss. In this
(0.25 mg/kg PO q24h) and aspirin (0.5 mg/kg PO q24h) case, treatment was continued for several months at the
were used to manage the protein-losing nephropathy. Two owner’s request while laboratory parameters and ultrasound
days after initiation of doxycycline treatment, the dog’s findings showed signs of progressive improvement. The
attitude and appetite improved. At discharge (day 9 of seropositivity to A. phagocytophilum may have reflected
hospitalization), the hematocrit was 26.6%, reticulocytes previous exposure to this organism, Anaplasma platys, or
150,000 cells/µL, platelet count 496,000/µL, BUN 31 mg/dL, serologic cross-reactivity between antibodies to E. canis
creatinine 1.5 mg/dL, albumin 1.8 g/dL, globulin 5 g/dL, and and the A. phagocytophilum antigen used in the test kit.

SUGGESTED READINGS 7. Felsheim RF, Kurtti TJ, Munderloh UG. Genome sequence of the
endosymbiont Rickettsia peacockii and comparison with virulent
Komnenou AA, Mylonakis ME, Kouti V, et al. Ocular manifestations Rickettsia rickettsii: identification of virulence factors. PLoS One.
of natural canine monocytic ehrlichiosis (Ehrlichia canis): a retrospec- 2009;4:e8361.
tive study of 90 cases. Vet Ophthalmol. 2007;10:137-142. 8. Harrus S, Perlman-Avrahami A, Mumcuoglu KY, et al. Molecular
Mylonakis ME, Koutinas AF, Breitschwerdt EB, et al. Chronic canine detection of Ehrlichia canis, Anaplasma bovis, Anaplasma platys,
ehrlichiosis (Ehrlichia canis): a retrospective study of 19 natural Candidatus Midichloria mitochondrii and Babesia canis vogeli in
cases. J Am Anim Hosp Assoc. 2004;40:174-184. ticks from Israel. Clin Microbiol Infect. 2011;17:459-463.
9. Masala G, Chisu V, Foxi C, et al. First detection of Ehrlichia canis
REFERENCES in Rhipicephalus bursa ticks in Sardinia, Italy. Ticks Tick Borne
1. Donatien A, Lestoquard F. Existence en Algerie d’une Rickettsia du Dis. 2012;3:396-397.
chien. Bull Soc Pathol Exot. 1935;28:418-419. 10. Wielinga PR, Gaasenbeek C, Fonville M, et  al. Longitudinal
2. Anderson BE, Greene CE, Jones DC, et  al. Ehrlichia ewingii sp. analysis of tick densities and Borrelia, Anaplasma, and Ehrlichia
nov., the etiologic agent of canine granulocytic ehrlichiosis. Int infections of Ixodes ricinus ticks in different habitat areas in The
J Syst Bacteriol. 1992;42:299-302. Netherlands. Appl Environ Microbiol. 2006;72:7594-7601.
3. Anderson BE, Dawson JE, Jones DC, et al. Ehrlichia chaffeensis, a 11. Satta G, Chisu V, Cabras P, et  al. Pathogens and symbionts in
new species associated with human ehrlichiosis. J Clin Microbiol. ticks: a survey on tick species distribution and presence of tick-
1991;29:2838-2842. transmitted micro-organisms in Sardinia. Italy. J Med Microbiol.
4. Hegarty BC, Maggi RG, Koskinen P, et al. Ehrlichia muris infection 2011;60:63-68.
in a dog from Minnesota. J Vet Intern Med. 2012;26:1217-1220. 12. Johnson EM, Ewing SA, Barker RW, et al. Experimental transmis-
5. Mavromatis K, Doyle CK, Lykidis A, et  al. The genome of the sion of Ehrlichia canis (Rickettsiales: Ehrlichieae) by Dermacentor
obligately intracellular bacterium Ehrlichia canis reveals themes variabilis (Acari: Ixodidae). Vet Parasitol. 1998;74:277-288.
of complex membrane structure and immune evasion strategies. 13. Beugnet F, Marie JL. Emerging arthropod-borne diseases of com-
J Bacteriol. 2006;188:4015-4023. panion animals in Europe. Vet Parasitol. 2009;163:298-305.
6. Dunning Hotopp JC, Lin M, Madupu R, et  al. Comparative 14. Groves MG, Dennis GL, Amyx HL, et al. Transmission of Ehrlichia
genomics of emerging human ehrlichiosis agents. PLoS Genet. canis to dogs by ticks (Rhipicephalus sanguineus). Am J Vet Res.
2006;2:e21. 1975;36:937-940.
288 SECTION 2  Bacterial Diseases

15. Harrus S, Kass PH, Klement E, et  al. Canine monocytic ehrlichi- 36. Mylonakis ME, Day MJ, Siarkou V, et al. Absence of myelofibrosis
osis: a retrospective study of 100 cases, and an epidemiological in dogs with myelosuppression induced by Ehrlichia canis infec-
investigation of prognostic indicators for the disease. Vet Rec. tion. J Comp Pathol. 2010;142:328-331.
1997;141:360-363. 37. Rikihisa Y, Ewing SA, Fox JC. Western immunoblot analysis of
16. Breitschwerdt EB, Abrams-Ogg AC, Lappin MR, et  al. Molecu- Ehrlichia chaffeensis, E. canis, or E. ewingii infections in dogs and
lar evidence supporting Ehrlichia canis-like infection in cats. J Vet humans. J Clin Microbiol. 1994;32:2107-2112.
Intern Med. 2002;16:642-649. 38. Seaman RL, Kania SA, Hegarty BC, et  al. Comparison of results
17. de Oliveira LS, Mourao LC, Oliveira KA, et al. Molecular detec- for serologic testing and a polymerase chain reaction assay to deter-
tion of Ehrlichia canis in cats in Brazil. Clin Microbiol Infect. mine the prevalence of stray dogs in eastern Tennessee seropositive
2009;15(suppl 2):53-54. to Ehrlichia canis. Am J Vet Res. 2004;65:1200-1203.
18. Braga Mdo S, André MR, Freschi CR, et al. Molecular and sero- 39. Gal A, Loeb E, Yisaschar-Mekuzas Y, et al. Detection of Ehrlichia
logical detection of Ehrlichia spp. in cats on São Luís Island, Mara- canis by PCR in different tissues obtained during necropsy from
nhão. Brazil. Rev Bras Parasitol Vet. 2012;21:37-41. dogs surveyed for naturally occurring canine monocytic ehrlichio-
19. Harrus S, Waner T, Keysary A, et al. Investigation of splenic func- sis. Vet J. 2008;175:212-217.
tions in canine monocytic ehrlichiosis. Vet Immunol Immuno- 40. de Castro MB, Machado RZ, de Aquino LP, et  al. Experimental
pathol. 1998;62:15-27. acute canine monocytic ehrlichiosis: clinicopathological and immu-
20. Brandao LP, Hasegawa MY, Hagiwara MK, et al. Platelet aggre- nopathological findings. Vet Parasitol. 2004;119:73-86.
gation studies in acute experimental canine ehrlichiosis. Vet Clin 41. Hildebrandt PK, Huxsoll DL, Walker JS, et  al. Pathology of
Pathol. 2006;35:78-81. canine ehrlichiosis (tropical canine pancytopenia). Am J Vet Res.
21. Cortese L, Terrazzano G, Piantedosi D, et  al. Prevalence of anti- 1973;34:1309-1320.
platelet antibodies in dogs naturally co-infected by Leishmania 42. Reardon MJ, Pierce KR. Acute experimental canine ehrlichiosis. I.
infantum and Ehrlichia canis. Vet J. 2011;188:118-121. Sequential reaction of the hemic and lymphoreticular systems. Vet
22. Mylonakis ME, Koutinas AF, Breitschwerdt EB, et  al. Chronic Pathol. 1981;18:48-61.
canine ehrlichiosis (Ehrlichia canis): a retrospective study of 19 43. Harrus S, Kenny M, Miara L, et al. Comparison of simultaneous
natural cases. J Am Anim Hosp Assoc. 2004;40:174-184. splenic sample PCR with blood sample PCR for diagnosis and
23. Gould DJ, Murphy K, Rudorf H, et al. Canine monocytic ehrlichio- treatment of experimental Ehrlichia canis infection. Antimicrob
sis presenting as acute blindness 36 months after importation into Agents Chemother. 2004;48:4488-4490.
the UK. J Small Anim Pract. 2000;41:263-265. 44. Schaefer JJ, Needham GR, Bremer WG, et al. Tick acquisition of
24. Komnenou AA, Mylonakis ME, Kouti V, et  al. Ocular mani- Ehrlichia canis from dogs treated with doxycycline hyclate. Antimi-
festations of natural canine monocytic ehrlichiosis (Ehrlichia crob Agents Chemother. 2007;51:3394-3396.
canis): a retrospective study of 90 cases. Vet Ophthalmol. 45. McClure JC, Crothers ML, Schaefer JJ, et al. Efficacy of a doxy-
2007;10:137-142. cycline treatment regimen initiated during three different phases
25. Buoro IBJ, Kanui TI, Atwell RB, et  al. Polymyositis associated of experimental ehrlichiosis. Antimicrob Agents Chemother.
with Ehrlichia canis infection in two dogs. J Small Anim Pract. 2010;54:5012-5020.
1990:31. 46. Shipov A, Klement E, Reuveni-Tager L, et al. Prognostic indicators
26. Dubey JP, Pimenta AL, Abboud LC, et al. Dermatitis in a dog asso- for canine monocytic ehrlichiosis. Vet Parasitol. 2008;153:131-138.
ciated with an unidentified Toxoplasma gondii-like parasite. Vet 47. Harrus S, Waner T, Neer TM. Ehrlichia and Anaplasma infections.
Parasitol. 2003;116:51-59. In: Greene CE, ed. Infectious Diseases of the Dog and Cat. 4th ed.
27. Hess PR, English RV, Hegarty BC, et  al. Experimental Ehrlichia St. Louis, MO: Elsevier Saunders; 2012:227-238.
canis infection in the dog does not cause immunosuppression. Vet 48. Matthewman LA, Kelly PJ, Brouqui P, et al. Further evidence for
Immunol Immunopathol. 2006;109:117-125. the efficacy of imidocarb dipropionate in the treatment of Ehrlichia
28. Neer TM, Breitschwerdt EB, Greene RT, et  al. Consensus state- canis infection. J S Afr Vet Assoc. 1994;65:104-107.
ment on ehrlichial disease of small animals from the infectious dis- 49. Eddlestone SM, Neer TM, Gaunt SD, et  al. Failure of imidocarb
ease study group of the ACVIM. American College of Veterinary dipropionate to clear experimentally induced Ehrlichia canis infec-
Internal Medicine. J Vet Intern Med. 2002;16:309-315. tion in dogs. J Vet Intern Med. 2006;20:840-844.
29. Leiva M, Naranjo C, Pena MT. Ocular signs of canine monocytic 50. Neer TM, Eddlestone SM, Gaunt SD, et al. Efficacy of enrofloxacin
ehrlichiosis: a retrospective study in dogs from Barcelona, Spain. for the treatment of experimentally induced Ehrlichia canis infec-
Vet Ophthalmol. 2005;8:387-393. tion. J Vet Intern Med. 1999;13:501-504.
30. Codner EC, Caceci T, Saunders GK, et al. Investigation of glomer- 51. Maurin M, Abergel C, Raoult D. DNA gyrase-mediated natural
ular lesions in dogs with acute experimentally induced Ehrlichia resistance to fluoroquinolones in Ehrlichia spp. Antimicrob Agents
canis infection. Am J Vet Res. 1992;53:2286-2291. Chemother. 2001;45:2098-2105.
31. Codner EC, Maslin WR. Investigation of renal protein loss in dogs 52. Aroch I, Harrus S. The use of hemopoietic growth factors: recom-
with acute experimentally induced Ehrlichia canis infection. Am binant human granulocyte colony stimulating factor and recombi-
J Vet Res. 1992;53:294-299. nant human erythropoietin in severe pancytopenia due to canine
32. Cortese L, Pelagalli A, Piantedosi D, et al. Platelet aggregation and monocytic ehrlichiosis. Israel J Vet Med. 2001;56:65-69.
haemostatic response in dogs naturally co-infected by Leishmania 53. Rudoler N, Baneth G, Eyal O, et al. Evaluation of an attenuated
infantum and Ehrlichia canis. J Vet Med A Physiol Pathol Clin strain of Ehrlichia canis as a vaccine for canine monocytic ehrlichi-
Med. 2006;53:546-548. osis. Vaccine. 2012;31:226-233.
33. Giudice E, Giannetto C, Gianesella M. Effect of desmopressin on 54. Tajima T, Rikihisa Y. Cytokine responses in dogs infected
immune-mediated haemorrhagic disorders due to canine mono- with Ehrlichia canis Oklahoma strain. Ann N Y Acad Sci.
cytic ehrlichiosis: a preliminary study. J Vet Pharmacol Ther. 2005;1063:429-432.
2010;33:610-614. 55. Unver A, Huang H, Rikihisa Y. Cytokine gene expression by
34. Meinkoth JH, Hoover JP, Cowell RL, et al. Ehrlichiosis in a dog peripheral blood leukocytes in dogs experimentally infected
with seizures and nonregenerative anemia. J Am Vet Med Assoc. with a new virulent strain of Ehrlichia canis. Ann N Y Acad Sci.
1989;195:1754-1755. 2006;1078:482-486.
35. Mylonakis ME, Koutinas AF, Leontides LS. Bone marrow mastocy- 56. Otranto D, Paradies P, Testini G, et al. Application of 10% imi-
tosis in dogs with myelosuppressive monocytic ehrlichiosis (Ehrlichia dacloprid/50% permethrin to prevent Ehrlichia canis exposure in
canis): a retrospective study. Vet Clin Pathol. 2006;35:311-314. dogs under natural conditions. Vet Parasitol. 2008;153:320-328.
CHAPTER 28  Ehrlichiosis 289

57. Stanneck D, Rass J, Radeloff I, et al. Evaluation of the long-term 70. Anziani OS, Ewing SA, Barker RW. Experimental transmission
efficacy and safety of an imidacloprid 10%/flumethrin 4.5% poly- of a granulocytic form of the tribe Ehrlichieae by Dermacentor
mer matrix collar (Seresto®) in dogs and cats naturally infested variabilis and Amblyomma americanum to dogs. Am J Vet Res.
with fleas and/or ticks in multicentre clinical field studies in Europe. 1990;51:929-931.
Parasit Vectors. 2012;5:66. 71. Yabsley MJ, Adams DS, O’Connor TP, et al. Experimental primary
58. Dantas-Torres F. The brown dog tick, Rhipicephalus sanguineus and secondary infections of domestic dogs with Ehrlichia ewingii.
(Latreille, 1806) (Acari: Ixodidae): from taxonomy to control. Vet Vet Microbiol. 2011;150:315-321.
Parasitol. 2008;152:173-185. 72. Goldman EE, Breitschwerdt EB, Grindem CB, et al. Granulocytic
59. Perez M, Bodor M, Zhang C, et al. Human infection with Ehrlichia ehrlichiosis in dogs from North Carolina and Virginia. J Vet Intern
canis accompanied by clinical signs in Venezuela. Ann N Y Acad Med. 1998;12:61-70.
Sci. 2006;1078:110-117. 73. Buller RS, Arens M, Hmiel SP, et  al. Ehrlichia ewingii, a
60. Ndip LM, Ndip RN, Esemu SN, et al. Ehrlichial infection in Cam- newly recognized agent of human ehrlichiosis. N Engl J Med.
eroonian canines by Ehrlichia canis and Ehrlichia ewingii. Vet 1999;341:148-155.
Microbiol. 2005;111:59-66. 74. Dumler JS, Madigan JE, Pusterla N, et al. Ehrlichioses in humans:
61. Oliveira LS, Oliveira KA, Mourao LC, et  al. First report of epidemiology, clinical presentation, diagnosis, and treatment. Clin
Ehrlichia ewingii detected by molecular investigation in dogs from Infect Dis. 2007;45(suppl 1):S45-S51.
Brazil. Clin Microbiol Infect. 2009;15(suppl 2):55-56. 75. Long SW, Zhang X, Zhang J, et  al. Evaluation of transovarial
62. Little SE, O’Connor TP, Hempstead J, et  al. Ehrlichia ewingii transmission and transmissibility of Ehrlichia chaffeensis (Rickettsiales:
infection and exposure rates in dogs from the south central United Anaplasmataceae) in Amblyomma americanum (Acari: Ixodidae). J
States. Vet Parasitol. 2010;172:355-360. Med Entomol. 2003;40:1000-1004.
63. Beall MJ, Alleman AR, Breitschwerdt EB, et al. Seroprevalence of 76. Breitschwerdt EB, Hegarty BC, Hancock SI. Sequential evaluation
Ehrlichia canis, Ehrlichia chaffeensis and Ehrlichia ewingii in dogs of dogs naturally infected with Ehrlichia canis, Ehrlichia chaffeen-
from North America. Parasit Vectors. 2012;5:29. sis, Ehrlichia equi, Ehrlichia ewingii, or Bartonella vinsonii. J Clin
64. Liddell AM, Stockham SL, Scott MA, et  al. Predominance Microbiol. 1998;36:2645-2651.
of Ehrlichia ewingii in Missouri dogs. J Clin Microbiol. 77. Zhang XF, Zhang JZ, Long SW, et  al. Experimental
2003;41:4617-4622. Ehrlichia chaffeensis infection in beagles. J Med Microbiol.
65. Goodman RA, Hawkins EC, Olby NJ, et al. Molecular identifica- 2003;52:1021-1026.
tion of Ehrlichia ewingii infection in dogs: 15 cases (1997-2001). 78. Kawahara M, Suto C, Rikihisa Y, et  al. Characterization of
J Am Vet Med Assoc. 2003;222:1102-1107. ehrlichial organisms isolated from a wild mouse. J Clin Microbiol.
66. Paddock CD, Yabsley MJ. Ecological havoc, the rise of white-tailed 1993;31:89-96.
deer, and the emergence of Amblyomma americanum-associated 79. Wen B, Rikihisa Y, Mott J, et  al. Ehrlichia muris sp. nov., iden-
zoonoses in the United States. Curr Top Microbiol Immunol. tified on the basis of 16S rRNA base sequences and serologi-
2007;315:289-324. cal, morphological, and biological characteristics. J Bacteriol.
67. Murphy GL, Ewing SA, Whitworth LC, et  al. A molecular 1995;45:250-254.
and serologic survey of Ehrlichia canis, E. chaffeensis, and 80. Pritt BS, Sloan LM, Johnson DK, et al. Emergence of a new patho-
E. ewingii in dogs and ticks from Oklahoma. Vet Parasitol. genic Ehrlichia species, Wisconsin and Minnesota, 2009. N Engl J Med.
1998;79:325-339. 2011;365:422-429.
68. Xiong Q, Bao W, Ge Y, et  al. Ehrlichia ewingii infection delays 81. Telford III SR, Goethert HK, Cunningham JA. Prevalence of
spontaneous neutrophil apoptosis through stabilization of mito- Ehrlichia muris in Wisconsin deer ticks collected during the mid
chondria. J Infect Dis. 2008;197:1110-1118. 1990s. Open Microbiol J. 2011;5:18-20.
69. Stockham SL, Tyler JW, Schmidt DA, et al. Experimental transmis-
sion of granulocytic ehrlichial organisms in dogs. Vet Clin Pathol.
1990;19:99-104.
CHAPTER 29

Anaplasmosis
Jane E. Sykes and Janet E. Foley

it forms host membrane-enclosed morulae. Ticks that belong to


Overview of Anaplasmosis the Ixodes ricinus-persulcatus complex are the major vectors for
First Described: First reports of Anaplasma phagocytophi- A. phagocytophilum. Anaplasma platys forms morulae within
lum came from sheep in 1951 (Scotland).1 Canine infec- platelets. The vector of A. platys is probably Rhipicephalus
tion was first reported in the United States (California) in ­sanguineus. As with the ehrlichioses, co-infections with other
1982.2 Anaplasma platys was first reported in the United pathogens that are transmitted by the same or other tick species
States (Florida) in 1978.3 may occur and influence the clinical manifestations of disease.
Cause: Anaplasma phagocytophilum (granulocytic anaplasmo-
sis), Anaplasma platys (thrombocytotropic anaplasmosis) Anaplasma phagocytophilum Infection
Affected Hosts: A. phagocytophilum causes disease in dogs,
cats, humans, ruminants (European strains), horses, and Etiology and Epidemiology
camelids. A. platys causes thrombocytopenia in dogs.
A. phagocytophilum causes granulocytic anaplasmosis in dogs,
Geographic Distribution: In the United States, A. phagocyto- humans, horses, and, in Europe, domestic ruminants that
philum is most prevalent in the upper Midwest, northeast include sheep, cattle, goats, and deer. Cats and camelids may
and western states. Infection also occurs throughout con- also be affected. A variety of wild animal species, which include
tinental Europe and the United Kingdom, Asia, and Russia. rodents and deer, act as reservoir hosts. For the midwestern and
The organism has been detected in dogs from Africa and eastern United States, white-footed mice (Peromyscus leuco-
South America. A. platys occurs throughout the Americas, pus) and eastern chipmunks may act as reservoirs, whereas in
Europe, Asia, Australia, the Middle East, and Africa. the western states, dusky-footed woodrats, gray squirrels, and
Mode of Transmission: Tick vectors, primarily Ixodes ricinus- chipmunks have been implicated. In Europe, bank voles, wood
persulcatus complex ticks transmit A. phagocytophilum. mice, shrews, and deer are likely reservoirs. Many strains of
Although unconfirmed, Rhipicephalus sanguineus is sus- A. phagocytophilum exist that differ in pathogenicity and host
pected to be the major vector of A. platys. tropism. For example, strains that infect domestic ruminants in
Europe and white-tailed deer in the United States appear to be
Major Clinical Signs: The major clinical signs of A. phagocy-
distinct from those that infect horses, humans, and dogs. Dogs,
tophilum infection are fever, lethargy, inappetence, and
cats, and humans are accidental hosts and are not important in
lameness due to polyarthritis, although vomiting, diar-
the transmission of infection to other host species.
rhea, cough, and neck pain may occur. A. platys usually
The geographic distribution of the disease follows that of the
causes no signs, but fever and lethargy are possible.
tick vectors (Figure 29-1). In North America, the tick vectors of
Differential Diagnoses: Major differential diagnoses include A. phagocytophilum are Ixodes scapularis in the northeastern
other tick-borne diseases (such as the ehrlichioses, rick- and upper midwestern states, and Ixodes pacificus in the West.
ettsioses, Lyme borreliosis and babesiosis), bartonellosis, In Europe, the primary vector is Ixodes ricinus, and the disease
leptospirosis, primary immune-mediated disease, and has been described in dogs throughout continental Europe and
lymphoma. in the UK. Ixodes persulcatus and Dermacentor silvarum ticks
Human Health Significance: A. phagocytophilum causes transmit the organism in Asia and Russia. Other Ixodes spp.
human granulocytic anaplasmosis. Dogs act as a sentinel ticks have also been implicated in transmission. Evidence of
for human infection and may carry unfed ticks to humans A. phagocytophilum infection has been found in dogs from
on their coats. Brazil and Tunisia, and a closely related organism was found
in dogs from South Africa.4-6 In humans, rare reports exist of
direct transmission that followed close contact with blood or
respiratory secretions, transplacental spread, or transmission

C
anine anaplasmosis is caused by Anaplasma through blood transfusion.
­phagocytophilum (formerly Ehrlichia equi, Ehrlichia Many seroprevalence studies of dogs have been reported
phagocytophila, and, in humans, the human granulo- worldwide.7 The seroprevalence varies with geographic loca-
cytic ehrlichiosis [HGE] agent) and Anaplasma platys. These are tion and whether the dogs studied were sick or healthy. The
tick-borne, gram-negative, obligately intracellular bacteria that prevalence of positive antibody titers in dogs from some regions
belong to the family Anaplasmataceae. A. phagocytophilum of Europe and North America exceeds 50%. A study that used a
predominantly infects neutrophils but also eosinophils, where commercially available ELISA assay to determine the prevalence

290
CHAPTER 29  Anaplasmosis 291

FIGURE 29-2  Life cycle of Ixodes scapularis ticks and Anaplasma phagocytophilum
infection. Uninfected larvae (top right) hatch in the late spring and acquire infection from
small rodents in the summer. They then over-winter (often in protected mouse burrows)
and then molt into nymphs the following spring. The nymphs feed in the late spring or
early summer on a variety of animal species including rodents, humans, deer, and dogs.
Nymphs molt into adults in the late summer to fall, which subsequently feed on large
mammals such as deer, where they mate and drop off. The females then lay eggs and die.
FIGURE 29-1  Distribution of Ixodes scapularis and Ixodes pacificus, the vectors of Dogs, cats and humans become infected by nymphs or adult ticks.
Anaplasma phagocytophilum, in the United States. The distribution of granulocytic ana-
plasmosis follows that of the tick vectors.
Clinical Features
of seroreactivity to A. phagocytophilum in more than 400,000
dogs from the United States revealed wide variation in seroprev- Signs and Their Pathogenesis
alences. Some counties in the upper Midwest and northeastern A. phagocytophilum is transmitted transstadially within the
United States had seroprevalences that exceeded 40%, although tick (i.e., from larva to nymph to adult), and not transovarially
the overall seroprevalence in these regions were 6.7% and 5.5%, (from adult to egg). Dogs and cats thus become infected after
respectively.8 Serologic cross-reactivity with A. platys may also exposure to infected nymphs or adult ticks, which acquire infec-
influence these data. tion when they feed on wild animal reservoir hosts as larvae
The seasonal pattern of disease reflects times of peak nymphal or nymphs (Figure 29-2). Ticks must attach for 36 to 48 hours
and adult tick activities, as well as periods when humans and for transmission to occur. Once A. phagocytophilum enters the
their dogs are active outdoors. In the western United States, bloodstream, it attaches to the sialylated ligands on the surface
A. phagocytophilum infection occurs most frequently in dogs of neutrophils, such as P-selectin glycoprotein ligand-1.7 The
between April and July, when nymphal ticks are abundant. organism then enters the neutrophil through caveolae-mediated
Some infections occur in October, during early questing of adult endocytosis. A. phagocytophilum survives in the harsh neutro-
ticks. In Minnesota and Wisconsin, most cases are diagnosed phil environment through dysregulation of neutrophil function
in late spring (May, June) and fall (October, November).9-11 and by bypassing phagolysosomal pathways. It inhibits neutro-
In a study from Berlin, most cases occurred between April and phil superoxide production and can reduce neutrophil motility
­September.12 The median age of clinically affected dogs is 6 to 8 and phagocytosis. A. phagocytophilum also reduces neutrophil
years (range, 6 months to 14 years).9,10,12-14 In the upper Mid- adherence to endothelium and inhibits neutrophil transmigra-
west of the United States, a bimodal age distribution has been tion into tissues, possibly through downregulation of selectin
recognized, with 25% of dogs 1 year of age or less, and 50% of molecule expression.7 This may promote its survival in periph-
dogs at least 8 years of age.10 Affected cats in the northeastern eral blood.
United States have ranged from 4 months to 13 years of age Normally, neutrophils circulate for 10 to 12 hours before
(mean, 3.7 years).15 Although no breed predispositions are rec- they enter tissues and undergo death through apoptosis.
ognized, in one study, golden retrievers comprised almost half A. phagocytophilum delays neutrophil apoptosis, which allows
of affected dogs; this may reflect the popularity of these dogs for it to survive longer periods of time within the neutrophil.7
outdoor activities.14 Infection with other tick-borne pathogens A. phagocytophilum may also infect other cell types, such as
is a risk factor for A. phagocytophilum infection. Co-infection bone marrow cells, endothelial cells, and megakaryocytes,
with Borrelia burgdorferi, the cause of Lyme disease, is com- although the importance of these cells in the pathogenesis of
mon because B. burgdorferi is transmitted by the same Ixodes infection is not clear.
tick species.8,10,11 In northern California, dogs seroreacted to The clinical signs and laboratory abnormalities that occur
A. phagocytophilum were 18 times more likely to be seroposi- in dogs and cats with granulocytic anaplasmosis probably
tive for Bartonella vinsonii subspecies berkhoffii than dogs that vary somewhat in different geographic locations as a result of
were seronegative.16 local strain variation. The vast majority of dogs infected with
292 SECTION 2  Bacterial Diseases

A. phagocytophilum show no clinical signs. Some dogs and cats on the infecting strain and host immune response to infection.
develop a self-limiting febrile illness, which in dogs occurs after In one study, treatment of dogs that had been experimentally
an incubation period of 1 to 2 weeks. Lethargy occurs in almost inoculated with A. phagocytophilum with prednisolone up to
all clinically affected cats and dogs. Fever and inappetence are 6 months after infection was followed by the development of
also common findings. Lameness, reluctance to move, polydip- positive PCR results for the organism and, in some dogs, throm-
sia, vomiting, diarrhea, and a soft cough can also occur. Gener- bocytopenia and reappearance of morulae on blood smears.23
alized lymphadenopathy and splenomegaly develop as a result One dog infected with a California strain of A. phagocytophi-
of reactive lymphoid hyperplasia and, in the spleen, concurrent lum was persistently PCR-positive through day 60 postinfec-
extramedullary hematopoiesis.17 Uncommonly, hemorrhage, tion, the last time point evaluated.23
manifested as mucosal petechiae, melena, or epistaxis, has been
reported in naturally infected dogs, but co-infections with other Physical Examination Findings
tick-borne pathogens may contribute to these signs in some The most common findings on physical examination in dogs
dogs.9,12,13,18 Neurologic signs such as seizures, circling, cervi- include lethargy, fever (up to 106.7°F [41.5°C]), dehydration,
cal pain, and decreased placing reactions have been described tachypnea, mild peripheral lymphadenopathy, and splenomeg-
in a few dogs from the upper Midwest with granulocytic ana- aly.7,10 Scleral injection may be noted. Lameness, reluctance to
plasmosis, but one dog with seizures had a history of idiopathic move, swollen joints, and pain on joint manipulation may also
epilepsy.9,18 Neurologic signs and detection of the organism in be detected in some dogs. Increased lung sounds, abdominal
the CSF have been uncommonly reported in humans.19 pain, epistaxis, petechial hemorrhages, and neurologic signs
Infection with A. phagocytophilum results in mild to mod- such as circling, cervical pain, and decreased placing reactions
erate thrombocytopenia, although other cytopenias may also occur uncommonly.
occur (Table 29-1). The mechanism(s) of these hematologic Physical examination findings reported in cats have been
abnormalities remain unclear. Anti-platelet antibodies occur similar to those described in dogs, and include fever, leth-
in serum from humans and dogs with granulocytic anaplasmo- argy, tachypnea, increased lung sounds, mild abdominal pain,
sis,12,20 and so immune-mediated mechanisms may contribute to hepatomegaly, splenomegaly, vomiting, ataxia, hyperesthesia,
thrombocytopenia. However, thrombocytopenia occurs in acute muscle and joint pain, lameness, conjunctivitis, and ocular
disease, before antibodies are noted, so other mechanisms may discharge.15,24,25
be important. The bone marrow of infected dogs shows mega-
karyocyte hyperplasia, so platelet destruction may be involved.21 Diagnosis
Impaired neutrophil function as a result of A. ­phagocytophilum
infection may predispose to development of secondary opportu- Granulocytic anaplasmosis should be suspected in dogs and
nistic infections or influence the outcome of co-infections with cats with acute febrile illness and thrombocytopenia that reside
other tick-borne pathogens such as B. burgdorferi. Although in endemic areas, regardless of tick exposure history. Diagno-
uncommon, opportunistic infections have been occasionally sis relies on detection of morulae within granulocytes, results
documented in humans and dogs with granulocytic anaplasmo- of acute and convalescent serology, or molecular testing using
sis and are well in small ruminants.22 PCR assays. The diagnostic criteria for confirmed human gran-
Infection with A. phagocytophilum may be self-limiting in ulocytic anaplasmosis are clinical signs and laboratory find-
dogs and cats, with minimal fatality or chronic disease mani- ings suggestive of granulocytic anaplasmosis together with (1)
festations. The extent to which A. phagocytophilum can persist detection of morulae within neutrophils combined with a single
in tissues and contribute to chronic disease manifestations in positive reciprocal antibody titer to A. phagocytophilum of
humans and dogs has been controversial, and may be dependent at least 80; (2) a fourfold increase or decrease in the antibody

TABLE 29-1
Hematologic Abnormalities in Nine Dogs with Granulocytic Anaplasmosis in Northern California*
Number ­below Number within Number above Range for dogs
the Reference the Reference the Reference with Granulocytic
Test Reference Range Range Range Range Anaplasmosis
Hematocrit (%) 40-55 6 3 0 19-45
Neutrophils (cells/µL) 3000-10,500 0 6 3 3513-18,592
Band neutrophils (cells/µL) Rare 0 1 8 0-1282
Monocytes (cells/µL) 150-1200 1 7 1 142-1346
Lymphocytes (cells/µL) 1,000-4000 7 2 0 71-2693
Eosinophils (cells/µL) 0-1500 0 9 0 0-328
Platelets (cells/µL)† 150,000-400,000 6 2 0 37,000-262,000

*Diagnosis was based on compatible clinical signs and either morulae within circulating neutrophils and/or a positive real-time PCR assay result for
A. phagocytophilum.
†Platelets were clumped for one dog.
CHAPTER 29  Anaplasmosis 293

titer within 4 weeks; (3) a positive PCR test result using specific Morulae appear as early as 4 days after experimental inocula-
A. phagocytophilum primers; or (4) isolation of A. ­phagocytophilum tion of dogs and persist for 4 to 8 days.17 The morulae are indis-
from blood.22 These criteria could also be applied to dogs. The use tinguishable from those of Ehrlichia ewingii, so serology or PCR
of multiple diagnostic modalities may be needed to confirm the assays are needed to confirm A. phagocytophilum infection.
diagnosis of granulocytic anaplasmosis in some dogs. In contrast to dogs, thrombocytopenia appears uncommon in
cats infected with A. phagocytophilum. None of 15 sick, PCR-
Laboratory Abnormalities positive cats from the northeastern United States were throm-
Complete Blood Count bocytopenic.15 The most common hematologic abnormality
Thrombocytopenia occurs in approximately 90% of dogs with in cats has been lymphopenia. Morulae have been detected in
granulocytic anaplasmosis.9,10,12,13 The platelet count in throm- some cats with granulocytic anaplasmosis.
bocytopenic dogs may occasionally be as low as 5000 platelets/
µL, although more often it is mild to moderately decreased (see Serum Biochemical Tests
Table 29-1).9,10,12-14 The majority of affected dogs are lympho- The most frequent serum biochemistry finding in dogs with
penic, but lymphocytosis can occur.9,12,13 Circulating reactive granulocytic anaplasmosis is mild to moderate hypoalbumin-
lymphocytes may be present. Anemia is common and typically emia (Table 29-2). Mild hyperglobulinemia, mild electrolyte
mild and nonregenerative. Both neutrophilia and neutropenia abnormalities (hypokalemia, hyponatremia, and metabolic aci-
occur, but most dogs have neutrophil counts that lie in the lower dosis), and a mild increase in the activities of serum ALP and to
half of the reference range. Low numbers of band neutrophils, a lesser extent ALT may occur.9,10,13,14
as well as mild neutrophil toxicity, are often present. Monocyto-
penia or monocytosis can occur.9,12,13 Cytologic examination of Urinalysis
blood smears often reveals morulae within granulocytes (Figure Urinalysis in dogs with granulocytic anaplasmosis may reveal
29-3). Morulae were detected in neutrophils from 36%, 56%, isosthenuria, hyposthenuria, and proteinuria. Urine protein-to-
67%, and 100% of dogs in four respective case series.9,12-14 creatinine ratios in two affected dogs from the United States were

A B C
FIGURE 29-3  Intracytoplasmic morula (A) and cocci (B) of Anaplasma phagocytophilum (arrows) within neutrophils of an 8-year-old male neutered golden retriever with granulocytic
anaplasmosis; in (C), morulae are present in neutrophils within the synovial fluid of a 5-year-old curly-coated retriever that had polyarthritis.

TABLE 29-2
Findings on Serum Biochemistry Analysis in Nine Dogs with Granulocytic Anaplasmosis in Northern California*
Number below Number within Number above Range for dogs
Reference the Reference the Reference the Reference with Granulocytic
Test Range Range Range Range Anaplasmosis
Sodium (mmol/L) 145-154 4 5 0 142-147
Potassium (mmol/L) 4.1-5.3 2 7 0 3.3-4.9
Bicarbonate (mmol/L) 16-26 3 6 0 11-18
Albumin (g/dL) 2.9-4.2 6 3 0 1.5-3.4
Globulin (g/dL) 2.3-4.4 0 8 1 2.7-6.0
Total bilirubin (mg/dL) 0-0.4 0 8 1 0-1.0
Alanine aminotransferase (U/L) 19-70 0 8 1 26-122
Alkaline phosphatase (U/L) 15-127 0 7 2 26-653

*Diagnosis was based on compatible clinical signs and either morulae within circulating neutrophils and/or a positive real-time PCR assay result for
A. phagocytophilum.
294 SECTION 2  Bacterial Diseases

1.5 and 2.2,10 but there is no evidence that A. ­phagocytophilum result, early in the course of illness, antibodies may be unde-
infection causes severe glomerulonephritis in dogs. tectable, so PCR assays may be more useful for diagnosis of
acute infection in the absence of detectable morulae. Because
Synovial Fluid Analysis positive titers can reflect previous exposure, demonstration of
Dogs with granulocytic anaplasmosis can develop neutrophilic a fourfold rise in titer is required. Antibody titers may persist
polyarthritis. Cytologic examination of synovial fluid in these for many months.14,23 In some human patients, antibody titers
dogs reveals increased numbers of nondegenerate neutrophils. have persisted as long as 3 years.22 In the United States, a silicon
In some affected dogs, morulae are found within these cells and disc-based ELISA assay is available commercially for detection
the synovial fluid can be PCR positive (Figure 29-3,C). of antibodies to A. ­phagocytophilum (Accuplex 4, Antech Diag-
nostics, Irvine, CA). The performance of this assay when com-
Diagnostic Imaging pared with IFA requires further study. An in-clinic lateral-flow
Plain Radiography ELISA device (IDEXX SNAP 4Dx Plus), which uses a recom-
Thoracic radiographs are usually normal in dogs with granu- binant Msp2/p44 protein, is also available for the detection of
locytic anaplasmosis but may show a mild interstitial infiltrate. antibodies to Anaplasma species in dog serum. Positive results
Focal alveolar infiltrates occur as well.26 obtained with these ELISA assays do not imply that A. phago-
cytophilum is the cause of illness, and as with IFA testing, nega-
Sonographic Findings tive results can occur in dogs with acute illness due to the lag in
Abdominal ultrasound may reveal splenomegaly, with a antibody production relative to the onset of clinical signs.
hypoechoic spleen, or mild abdominal lymphadenopathy. Serologic cross-reactivity between Anaplasma species occurs.
Dogs infected with A. platys also seroreact to A. ­phagocytophilum
Microbiologic Tests antigen; this includes the recombinant Msp2 assay.8 Serologic
Diagnostic assays available for anaplasmosis in dogs and cats cross-reactivity between A. ­phagocytophilum and E. canis also
are shown in Table 29-3. occurs, but is relatively uncommon and minor.13,26,27 Dogs diag-
nosed with granulocytic anaplasmosis that have antibodies to
Serologic Diagnosis A. ­phagocytophilum generally lack antibodies to E. canis using
Diagnosis of granulocytic anaplasmosis can be accomplished IFA testing.7,10
with acute and convalescent serology. Many veterinary laborato-
ries perform serologic testing using immunofluorescent antibody Molecular Diagnosis Using the Polymerase Chain Reaction
(IFA) techniques. IgG antibodies are first detectable approximately Several conventional and real-time PCR assays for
8 days after exposure, 2 to 5 days after morulae appear. As a A. ­phagocytophilum exist for detection of A. phagocytophilum

TABLE 29-3
Diagnostic Assays Available for Anaplasmosis in Dogs and Cats
Assay Specimen Type Target Performance
Cell culture Whole blood Anaplasma phagocytophilum; Not widely offered or utilized for routine diagnostic
Anaplasma platys cannot be purposes. Requires several weeks’ incubation.
cultured
Cytology Whole blood, buffy coat A. phagocytophilum or Low sensitivity (especially for A. platys infection).
smears, body fluids, A. platys morulae Morulae of A. phagocytophilum cannot be
­tissue aspirates ­distinguished from those of Ehrlichia ewingii.
Morulae may be confused with cytoplasmic
­granules or stain precipitate.
IFA serology Serum Antibodies to Anaplasma spp. Acute and convalescent serology is required for
diagnosis of acute infection, because initial results
may be negative in dogs with acute disease and
positive results may reflect previous exposure
rather than active infection. Cross-reactivity
­occurs between Anaplasma spp.
ELISA serology Serum Antibodies to Anaplasma spp. Rapid, inexpensive, can be performed as an in-practice
antigens test. Similar limitations as IFA. Lack of quantitation
limits ability to document seroconversion.
PCR Whole blood, splenic or A. phagocytophilum or Confirms active infection. Sensitivity and specificity
lymph node aspirates, A. platys DNA may vary depending on assay design and speci-
bone marrow aspirates, men type. Because healthy animals may be PCR
buffy coat specimens, positive, positive PCR results must be interpreted
tissue specimens in light of the clinical signs.

IFA, Immunofluorescent antibody.


CHAPTER 29  Anaplasmosis 295

DNA in peripheral blood, buffy coat, bone marrow, or splenic to document whether persistence occurs through similar mecha-
tissue. These can be useful for early diagnosis of granulocytic nisms in other host species. It is possible that recovery from nat-
anaplasmosis in dogs and cats. The sensitivity and specificity ural infection confers long-term protection against development
of PCR assays may vary depending on the assay design and of disease. Reinfection has not been reported in dogs, but was
the laboratory used. Most, but not all, assays detect either described in one human patient.36 A vaccine for granulocytic
the 16S rRNA gene or the outer surface protein gene msp2 anaplasmosis is not available for companion animals.
(p44). Assays that detect the msp2 gene are usually specific for
A. ­phagocytophilum, whereas assays that detect the 16S rRNA Prevention
gene may detect other Anaplasma species, and even other bacteria.
A. phagocytophilum DNA has occasionally been amplified from Infection may be prevented by avoidance of tick exposure,
healthy dogs, so results must be interpreted in light of the clini- prompt tick removal, and use of topical ectoparasiticides (see
cal signs.11 In experimentally infected dogs, whole-blood PCR Chapter 28). Although not a guaranteed protection, combina-
becomes positive several days before and after morulae appear tions of topical imidacloprid and permethrin, or fipronil, amitraz
on blood smears.17,23 and (S)-methoprene, prevent transmission of A. phagocytophi-
lum to dogs from infected ticks.37,37a
Culture
A. phagocytophilum can be isolated in human promyelocytic Public Health Aspects
leukemia cell lines (HL-60) and tick embryo cell lines. Culture
is highly sensitive for diagnosis of acute infection in human A. phagocytophilum infection was first recognized in humans
patients,28 but it is not routinely used in dogs for diagnostic in the upper Midwest of the United States in the early 1990s
purposes because it requires special facilities, technical exper- and has since been increasingly recognized in humans from the
tise, and prolonged incubation. United States, Europe, and Asia. Human granulocytic anaplas-
mosis closely resembles the disease in dogs, which has been
Pathologic Findings described as an “influenza-like illness after a tick bite.”38 Men
Little information is available regarding the pathology of gran- are affected slightly more frequently than women. In Europe,
ulocytic anaplasmosis in dogs. Tissue injury appears to result the disease is most commonly reported in from Sweden and
from the host inflammatory response, rather than the bacterial Slovenia, where it was first reported in 1997.39 Morulae are
infection itself.29 In people, splenic lymphoid depletion, mac- observed less often in humans with granulocytic anaplasmosis
rophage aggregates and apoptosis within the liver, paracortical in Europe, and the disease may be milder than that described in
lymphoid hyperplasia, and hemophagocytic cells within reticu- humans from the United States.
loendothelial tissues have been described.30 The most common clinical signs reported in human patients
are myalgia, headache (which is often severe), malaise, and chills.
Treatment and Prognosis Anorexia, nausea, arthralgias, and cough may also occur.22,38,40
The disease typically resolves within 2 months in the absence of
The treatment of choice for granulocytic anaplasmosis in dogs appropriate antibiotic treatment. Occasionally more severe dis-
is doxycycline (5 mg/kg PO q12h). The optimum duration ease may occur. In one study, up to 17% of affected humans
is unknown, but 2 weeks may be sufficient. The prognosis is required admission to an intensive care unit.38 Death occurs in
excellent. Most dogs show clinical improvement within 24 to 1% or fewer clinically affected humans, usually as a result of
48 hours of treatment, although some dogs require more than complications such as a septic or toxic shock–like syndrome,
1 week of treatment before clinical signs resolve.10,13 Platelet respiratory insufficiency, opportunistic fungal or viral infections,
counts normalize 2 to 14 days after treatment is initiated.10 In rhabdomyolysis, acute renal failure, hemorrhage, or neurologic
one study, 30% of 23 owners reported that their dog returned disease.22 Severe illness tends to occur in humans of advanced age
to normal activities in 1 to 2 days after treatment was initiated, or with concurrent immunosuppressive illness or drug therapy.
30% reported this took 3 to 5 days, 21% 1 to 3 weeks, and Laboratory testing of peripheral blood typically reveals normal or
17% reported that it took a month or longer for their dogs to slightly decreased white blood cell and platelet counts, sometimes
return to normal activity.10 Death due to granulocytic anaplas- with a neutrophilic left shift, and mild to moderate elevation of
mosis has not been described in dogs. Intravenous crystalloid hepatic transaminase activities. In the United States, the disease
fluids and antiemetics may be required for supportive care until is reportable to the Centers for Disease Control and Prevention
clinical signs improve. (CDC). Dogs act as sentinels for human exposure and may be a
source of infection through mechanical carriage of infected ticks
Immunity and Vaccination to humans. Blood from affected dogs should be handled with
caution, and needle-stick injuries should be avoided.
The immune response to A. phagocytophilum infection is not
fully characterized. Both humoral and cell-mediated immune Anaplasma platys Infection
responses appear to be important in clearance of infection. Host
cytokines such as IFN-γ may play a role in the initial control Etiologic Agent and Epidemiology
of A. phagocytophilum infection31,32 but may also contribute to
the inflammatory process associated with disease.33 In lambs, Anaplasma platys infects and forms inclusions within platelets
A. phagocytophilum evades the host immune response through and is the cause of canine cyclic thrombocytopenia, or thrombo-
differential expression of MSP2, an outer surface protein cytotropic anaplasmosis. It does not appear to infect cats. The
involved in immune recognition.34 Variation in MSP2 also occurs organism is very widespread, and infections occur throughout
in chronically infected woodrats.35 Further research is required the Americas, Europe, Asia, Australia, the Middle East, and
296 SECTION 2  Bacterial Diseases

Africa. R. sanguineus ticks are believed to transmit A. platys, routine diagnostic purposes. Molecular diagnostic assays have
because the DNA of A. platys has frequently been found in overcome the need for direct fluorescent antibody or immuno-
R. sanguineus ticks worldwide, and dogs infected with A. platys cytochemical staining.
are often co-infected with E. canis. However, in a single attempt,
experimental transmission of A. platys by R. sanguineus ticks Serologic Diagnosis
failed.41 A. platys DNA has been found in other tick species, Recent infection with an Anaplasma species can be detected
such as Dermacentor auratus ticks in Thailand,42 ­Rhipicephalus through the use of acute and convalescent serology with IFA.
turanicus ticks from Israel,43 and Haemaphysalis spp. and Because antibodies to A. phagocytophilum bind to A. platys
Ixodes nipponensis ticks from Korea.44 Different strains of antigens it is not possible to distinguish the immune response
A. platys exist that appear to vary in pathogenicity. The organ- to each of these pathogens with currently available assays. This
ism has never been isolated in cell culture. can be problematic in geographic regions where both A. platys
and A. phagocytophilum are found. A positive result on com-
Clinical Features mercially available ELISA assays also only indicates previous
exposure to an Anaplasma species. Dogs with recent infection
Signs and Their Pathogenesis may be seronegative, because antibodies to A. platys are not
Anaplasma platys causes thrombocytopenia in dogs, most often detectable for 1 to 2 weeks after inoculation. Because a single
in the absence of other clinical signs, although fever, lethargy, positive test result only indicates previous exposure, and sero-
lymphadenopathy, uveitis, pallor, and mucosal hemorrhages have prevalence rates in endemic areas are high, thrombocytopenia
been described.45,46 Co-infections with other vector-borne patho- in a seropositive dog may be due to an etiology other than
gens may contribute to clinical signs in some dogs. Thrombocy- ­Anaplasma infection. Cross-reactivity to E. canis antigens does
topenia occurs 1 to 2 weeks after experimental inoculation. This not occur in dogs with A. platys infections, so positive titers to
initial episode is associated with the highest number of organisms E. canis and A. platys may represent previous exposure to both
in platelets, as detected using light microscopic examination of pathogens, or to other Ehrlichia or Anaplasma species.
stained blood smears.47 The platelet count nadir occurs 2 to 3
weeks postinfection and in some dogs may be lower than 20,000 Molecular Diagnosis Using the Polymerase Chain Reaction
platelets/µL. Visible organisms then disappear from platelets, and Several conventional and real-time PCR assays have been
the platelet count returns to normal or near-normal limits within described for detection of A. platys DNA. In the absence of
3 to 4 days. This also corresponds with a decrease in organism clearly identifiable morulae within platelets, molecular diag-
load and failure to detect the organism in peripheral blood with nostic testing with reliable PCR assays that specifically detect
real-time PCR, although bone marrow and splenic aspirates may A. platys DNA (as opposed to A. phagocytophilum DNA or
remain positive.48 Cycles of thrombocytopenia and bacteremia Anaplasma species DNA) is the only way to confirm active
then occur at intervals of 7 to 14 days, after which infection and infection. Real-time PCR assays that detect A. platys DNA
thrombocytopenia persist in some dogs but morulae or inclusions are available commercially and in combination with assays for
become more difficult to detect within platelets.47 The mecha- other vector-borne pathogens. Suitable specimens include whole
nism of thrombocytopenia is unknown, but direct damage to blood, buffy coats, or bone marrow and splenic aspirates,48
platelets, sequestration of platelets in the spleen, and immune- although it is not clear which of these specimens is optimal.
mediated destruction have been hypothesized to play a role. When buffy coats were used, PCR assays for A. platys became
Co-infection of dogs with A. platys and E. canis may lead positive as early 3 to 5 days after experimental infection, but
to more severe anemia than occurs with isolated E. canis by day 21, negative results occurred in some dogs.48,49 The use
infection.49 The duration of A. platys infection may also be of splenic or bone marrow aspirates may yield positive results
prolonged. when whole blood or buffy coat PCR for A. platys is negative
yet infection is still suspected.
Physical Examination Findings
Most dogs with A. platys infection show no clinical signs. Fever, Pathologic Findings
petechial hemorrhages, and uveitis have been reported, but co- Pathologic findings have been described in a small number
infections with other vector-borne pathogens may have contrib- of dogs that were experimentally infected with A. platys, and
uted to these signs. necropsied early in the course of infection.50 The only gross
necropsy finding was generalized lymphadenomegaly. Lesions
Diagnosis consisted of reactive lymphoid hyperplasia and erythrophago-
cytosis by sinusoidal macrophages within lymph nodes and the
Laboratory Abnormalities spleen, crescent-shaped regions of perifollicular hemorrhage in
Usually, the only abnormality present on the CBC, biochemistry the spleen, mild lymphoplasmacytic infiltrates in the renal inter-
panel, and urinalysis in dogs with A. platys infection is throm- stitium, and multifocal hyperplasia of Kupffer’s cells in the liver.
bocytopenia. A mild, nonregenerative anemia may be found. Megakaryocyte numbers in the bone marrow were increased in
The diagnosis can be made when morulae are seen within plate- some dogs.
lets on blood smears, but this is insensitive, and other inclusions
or stain precipitate may be mistaken for organisms. Treatment and Prognosis
Microbiologic Tests The recommended treatment for thrombocytopenic dogs
Direct Fluorescent Antibody infected with A. platys is doxycycline. The optimum dose and
Direct fluorescent antibody techniques can be used to identify duration of treatment is unknown, but it is apparently eliminated
A. platys in platelets, but these are not widely available for using regimens effective for treatment of E. canis infection.
CHAPTER 29  Anaplasmosis 297

Infection could not be detected with PCR for a 3-week follow- Prevention
up period after just 8 days of doxycycline treatment (10 mg/kg
PO q24h), which was initiated in the acute phase of infection.51 See the previous discussion of Anaplasma phagocytophilum
However, one dog remained infected after 2 weeks of treatment infection.
with tetracycline. In another study, doxycycline treatment was
administered for 28 days, and infection could not be detected Public Health Aspects
after this time point, even after pharmacologic immunosuppres-
sion with dexamethasone several months later.49 Whether the Infection with A. platys has not been confirmed in humans.
duration of infection at the time of treatment affects treatment
efficacy is unknown.

CASE EXAMPLE Eyes, Ears, Nose, and Throat: Mild gingivitis and dental
calculus were present. No other significant abnormalities
were noted.
Signalment: “Copper,” an 8-year old male neutered Labrador Musculoskeletal: Body condition score 4/9. The dog was
retriever from Napa, CA ambulatory but had a slightly stiff thoracic limb gait. A
History: Copper was brought to the University of California, decreased range of motion of coxofemoral joints was
Davis, VMTH emergency service for the problems of lethargy, detected, with the left worst than the right. There was no
inappetence, apparent neck pain of 1 week’s duration, and evidence of joint pain or effusion.
fever. The dog had been taken to a local veterinary clinic on Cardiovascular, Respiratory Systems, Gastrointestinal
the day the illness began, and thrombocytopenia (113,000 and Urogenital Systems: No clinically significant findings
platelets/µL) was detected on a CBC. ELISA serology for were identified. Rectal examination revealed no significant
antibodies to E. canis, B. burgdorferi, and A. phagocytophilum abnormalities.
and Dirofilaria immitis antigen (4Dx SNAP test, IDEXX Lymph Nodes: All peripheral lymph nodes were normal sized
Laboratories) was negative. Treatment with enrofloxacin but slightly firm on palpation.
(2.4 mg/kg PO q12h) and metronidazole was instituted, Neurologic Examination: Mentation and cranial nerve
during which time the dog improved clinically, but when function were within normal limits. The dog was reluctant to
the metronidazole was discontinued after 5 days, lethargy, move his neck on manipulation, especially to the right side,
inappetence, and signs of neck pain returned 2 days and palpation of the cervical spine elicited pain. Decreased
later. A CBC at that time showed no clinically significant placing reactions were noted in the left pelvic limb.
abnormalities. Treatment with enrofloxacin was continued, Laboratory Findings:
and methocarbamol (11.6 mg/kg PO q8h) and meloxicam CBC:
(0.1 mg/kg PO q24h) were instituted for the neck pain. The HCT 35.8% (40%-55%)
owner took Copper’s rectal temperature at home on the MCV 64 fL (65-75 fL)
morning he was brought to the VMTH, and it was 105.6°F MCHC 35.8 g/dL (33-36 g/dL)
(40.9°C). Copper lived on a 20-acre winery with a reservoir Reticulocyte count 10,200 cells/µL (7000-65,000 cells/µL)
and had access to wildlife and frequent tick exposure. There WBC 7100 cells/µL (6000-13,000 cells/µL)
had been no known exposure to toxins and no travel history. Neutrophils 6603 cells/µL (3000-10,500 cells/µL)
The dog received monthly heartworm preventative, and flea Band neutrophils 284 cells/µL
and tick preventative was used, but only when the owner Lymphocytes 71 cells/µL (1000-4000 cells/µL)
found ticks. He was normally fed a commercially available Monocytes 142 cells/µL (150-1200 cells/µL)
dry dog food. Platelets 37,000/µL (150,000-400,000 platelets/µL)
Current Medications: Enrofloxacin (2.4 mg/kg PO q12h), MPV 17.2 fL (7-13 fL).
methocarbamol (11.6 mg/kg PO q8h), and meloxicam Slight toxicity was detected in neutrophils and band neutro-
(0.1 mg/kg PO q24h). phils, and multiple neutrophils contained basophilic intra-
Other Medical History: Nine months previously, Copper cytoplasmic inclusions (morulae) (see Figure 29-3).
was seen by the local veterinary clinic for neck pain. The Serum Chemistry Profile:
dog was treated with methocarbamol, a nonsteroidal Anion gap 23 mmol/L (10-24 mmol/L)
antiinflammatory drug, and rest, and the pain resolved after Sodium 145 mmol/L (145-154 mmol/L)
7 days. The owners also reported that Copper had exhibited Potassium 3.7 mmol/L (3.6-5.3 mmol/L)
some weakness of his left pelvic limb for several years. Chloride 115 mmol/L (108-118 mmol/L)
Physical Examination: Bicarbonate 11 mmol/L (16-26 mmol/L)
Body weight: 43.1 kg Phosphorus 3.9 mg/dL (3.0-6.2 mg/dL)
General: Quiet, alert, responsive, adequately hydrated. Calcium 10.2 mg/dL (9.7-11.5 mg/dl)
T = 103.7°F (39.8°C), HR = 110 beats/min, panting, mucous BUN 12 mg/dL ( 5-21 mg/dL)
membranes pink, CRT <2 s. Creatinine 0.7 mg/dL (0.3-1.2 mg/dL)
Integument: No clinically significant abnormalities were Glucose 61 mg/dL (64-123 mg/dL)
identified. No ectoparasites were noted. Total protein 6.8 g/dL (5.4-7.6 g/dL)
298 SECTION 2  Bacterial Diseases

Albumin 3.1 g/dL (3.0-4.4 g/dL) IFA serology for vector-borne pathogens: Positive for antibody
Globulin 3.7 g/dL (1.8-3.9 g/dL) to A. phagocytophilum (1:2560). Negative for antibodies to
ALT 54 U/L (19-67 U/L) R. rickettsia at <1:40. Negative for antibodies to Babesia canis
AST 26 U/L (19-42 U/L) at <1:40. Weak positive for antibody to E. canis at 1:40.
ALP 106 U/L (21-170 U/L) Vector-borne real-time PCR panel (whole blood): Positive for
Creatine kinase 79 U/L (51-399 U/L) A. phagocytophilum (Msp gene PCR). Negative for Ana-
GGT < 3 U/L (0-6 U/L) plasma platys, Bartonella spp., Borrelia burgdorferi, Ehrlichia
Cholesterol 255 mg/dL (135-361 mg/dL) canis, and Rickettsia spp. DNA.
Total bilirubin 0.1 mg/dL (0-0.2 mg/dL) Diagnosis: Granulocytic anaplasmosis
Magnesium 2.1 mg/dL (1.5-2.6 mg/dL). Treatment: Copper was treated with doxycycline (5 mg/
Urinalysis: SGr 1.031; pH 8.0, 25 mg/dL protein, no bilirubin, kg PO q12h) the day after admission, when results of the
25 erythrocytes/µL hemoprotein, no glucose, 0-3 WBC/HPF, CBC were available. In the interim, pain was managed with
15-25 RBC/HPF, no other significant abnormalities were meloxicam and hydromorphone, and IV crystalloid fluids
detected. were administered. A day later, the fever had resolved
Imaging Findings: and the dog’s appetite and neck pain had improved, but
Spinal Radiographs: Multiple right lateral projections of thrombocytopenia persisted (36,000 platelets/µL). Two
the spine were reviewed. There were multiple sites of days after initiation of doxycycline treatment, the platelet
mild ventral spondylosis deformans throughout the count was 302,000 platelets/µL, and all other hematologic
thoracolumbar spine. Severe spondylosis was seen at the variables that were abnormal had returned to the reference
lumbosacral space with sclerosis of the endplates either range. At a recheck examination, 2 weeks after discharge,
side, most likely due to degenerative change. There was Copper was doing well, with no evidence of pain or stiffness.
severe osseous remodeling of the lumbar articular facets, A CBC showed a platelet count of 487,000 platelets/µL.
which was consistent with osteoarthrosis. Small areas of Comments: This was an unusual case of granulocytic
intervertebral disc mineralization were seen at the L2-3 and anaplasmosis in a dog that developed apparent cervical
L4-5 spaces. pain in association with infection. The dog also had
Thoracic Radiographs: The cardiovascular and pulmonary osteoarthritis, but clinical signs resolved almost completely
structures were within normal limits. There was slightly once doxycycline was instituted. Although the dog’s joints
increased soft tissue opacity in the region of the sternal did not appear painful or swollen on physical examination,
lymph node on the right lateral projection. it is possible that polyarthritis may have contributed to the
Abdominal Ultrasound: The liver was mildly hypoechoic but clinical signs of stiffness. Serology was initially negative
normal in size. The spleen was markedly enlarged but had because infection was too recent for an antibody response to
normal echogenicity. The rest of the abdominal organs were have developed. The diagnosis of granulocytic anaplasmosis
within normal limits. was confirmed with molecular diagnostic testing and
Microbiologic Testing: Aerobic bacterial urine culture: No supported by seroconversion to A. phagocytophilum and
growth identification of morulae within neutrophils. The weak
ELISA serology for vector-borne pathogens (IDEXX 4Dx SNAP positive IFA test result to E. canis may have reflected serologic
test): Positive for antibodies to A. phagocytophilum. Nega- cross-reactivity. Treatment with enrofloxacin was apparently
tive for antibodies to E. canis and B. burgdorferi. Negative for ineffective. The need for more consistent application of tick
D. immitis antigen. preventatives was discussed with the owners.

SUGGESTED READING 6. Santos HA, Pires MS, Vilela JA, et  al. Detection of Anaplasma
phagocytophilum in Brazilian dogs by real-time polymerase chain
Carrade DD, Foley JE, Borjesson DL, et  al. Canine granulocytic ana- reaction. J Vet Diagn Invest. 2011;23:770-774.
plasmosis: a review. J Vet Intern Med. 2009;23:1129-1141. 7. Carrade DD, Foley JE, Borjesson DL, et  al. Canine granulocytic
anaplasmosis: a review. J Vet Intern Med. 2009;23:1129-1141.
REFERENCES 8. Bowman D, Little SE, Lorentzen L, et al. Prevalence and geographic
1. Foggie A. Studies on the infectious agent of tick-borne fever in distribution of Dirofilaria immitis, Borrelia burgdorferi, Ehrlichia
sheep. J Pathol Bacteriol. 1951;63:1-15. canis, and Anaplasma phagocytophilum in dogs in the United
2. Madewell BR, Gribble DH. Infection in two dogs with States: results of a national clinic-based serologic survey. Vet Para-
an agent resembling Ehrlichia equi. J Am Vet Med Assoc. sitol. 2009;160:138-148.
1982;180:512-514. 9. Greig B, Asanovich KM, Armstrong PJ, et  al. Geographic, clini-
3. Harvey JW, Simpson CF, Gaskin JM. Cyclic thrombocyto- cal, serologic, and molecular evidence of granulocytic ehrlichiosis,
penia induced by a Rickettsia-like agent in dogs. J Infect Dis. a likely zoonotic disease, in Minnesota and Wisconsin dogs. J Clin
1978;137:182-188. Microbiol. 1996;34:44-48.
4. M’Ghirbi Y, Ghorbel A, Amouri M, et  al. Clinical, serological, 10. Granick JL, Armstrong PJ, Bender JB. Anaplasma phagocytophi-
and molecular evidence of ehrlichiosis and anaplasmosis in dogs in lum infection in dogs: 34 cases (2000-2007). J Am Vet Med Assoc.
Tunisia. Parasitol Res. 2009;104:767-774. 2009;234:1559-1565.
5. Inokuma H, Oyamada M, Kelly PJ, et  al. Molecular detec- 11. Beall MJ, Chandrashekar R, Eberts MD, et  al. Serological and
tion of a new Anaplasma species closely related to Anaplasma molecular prevalence of Borrelia burgdorferi, Anaplasma phago-
­phagocytophilum in canine blood from South Africa. J Clin Micro- cytophilum, and Ehrlichia species in dogs from Minnesota. Vector
biol. 2005;43:2934-2937. Borne Zoonotic Dis. 2008;8:455-464.
CHAPTER 29  Anaplasmosis 299

12. Kohn B, Galke D, Beelitz P, et al. Clinical features of canine gran- 33. Martin ME, Caspersen K, Dumler JS. Immunopathology and
ulocytic anaplasmosis in 18 naturally infected dogs. J Vet Intern ehrlichial propagation are regulated by interferon-gamma and
Med. 2008;22:1289-1295. interleukin-10 in a murine model of human granulocytic ehrlichio-
13. Poitout FM, Shinozaki JK, Stockwell PJ, et al. Genetic variants of sis. Am J Pathol. 2001;158:1881-1888.
Anaplasma phagocytophilum infecting dogs in western Washing- 34. Granquist EG, Stuen S, Lundgren AM, et al. Outer membrane pro-
ton state. J Clin Microbiol. 2005;43:796-801. tein sequence variation in lambs experimentally infected with Ana-
14. Egenvall AE, Hedhammar AA, Bjoersdorff AI. Clinical features and plasma phagocytophilum. Infect Immun. 2008;76:120-126.
serology of 14 dogs affected by granulocytic ehrlichiosis in Sweden. 35. Rejmanek D, Foley P, Barbet A, et al. Antigen variability in Ana-
Vet Rec. 1997;140:222-226. plasma phagocytophilum during chronic infection of a reservoir
15. Savidge C, Ewing P, Andrews J, et  al. Anaplasma phagocytophi- host. Microbiology. 2012;158:2632-2641.
lum in cats: a retrospective analysis and clinical evaluation of fif- 36. Horowitz HW, Aguero-Rosenfeld M, Dumler JS, et al. Reinfection
teen cases from the north eastern United States. J Vet Intern Med. with the agent of human granulocytic ehrlichiosis. Ann Intern Med.
2011;25:706:(abst). 1998;129:461-463.
16. Foley JE, Brown RN, Gabriel MW, et  al. Spatial analysis of the 37. Blagburn BL, Spencer JA, Billeter SA, et  al. Use of imidacloprid-
exposure of dogs in rural north-coastal California to vectorborne permethrin to prevent transmission of Anaplasma phagocytophi-
pathogens. Vet Rec. 2007;161:653-657. lum from naturally infected Ixodes scapularis ticks to dogs. Vet
17. Egenvall A, Bjoersdorff A, Lilliehook I, et al. Early manifestations Ther. 2004;5:212-217.
of granulocytic ehrlichiosis in dogs inoculated experimentally with 37a. McCall JW, Baker CF, Mather TN, et  al. The ability of a topi-
a Swedish Ehrlichia species isolate. Vet Rec. 1998;143:412-417. cal novel combination of fipronil, amitraz and (S)-methoprene to
18. Eberts MD, Vissotto de Paiva Diniz PP, Beall MJ, et al. Typical and protect dogs from Borrelia burgdorferi and Anaplasma phagocyto-
atypical manifestations of Anaplasma phagocytophilum infection philum infections transmitted by Ixodes scapularis. Vet Parasitol.
in dogs. J Am Anim Hosp Assoc. 2011;47:e86-e94. 2011;179:335-342.
19. Lee FS, Chu FK, Tackley M, et al. Human granulocytic ehrlichiosis 38. Bakken JS, Krueth J, Wilson-Nordskog C, et al. Clinical and labo-
presenting as facial diplegia in a 42-year-old woman. Clin Infect ratory characteristics of human granulocytic ehrlichiosis. JAMA.
Dis. 2000;31:1288-1291. 1996;275:199-205.
20. Wong SJ, Thomas JA. Cytoplasmic, nuclear, and platelet autoan- 39. Strle F. Human granulocytic ehrlichiosis in Europe. Int J Med
tibodies in human granulocytic ehrlichiosis patients. J Clin Micro- Microbiol. 2004;293(suppl 37):27-35.
biol. 1998;36:1959-1963. 40. Dumler JS, Madigan JE, Pusterla N, et al. Ehrlichioses in humans:
21. Lilliehook I, Egenvall A, Tvedten HW. Hematopathology in dogs epidemiology, clinical presentation, diagnosis, and treatment. Clin
experimentally infected with a Swedish granulocytic Ehrlichia spe- Infect Dis. 2007;45(suppl 1):S45-S51.
cies. Vet Clin Pathol. 1998;27:116-122. 41. Simpson RM, Gaunt SD, Hair JA, et al. Evaluation of Rhipicepha-
22. Bakken JS, Dumler S. Human granulocytic anaplasmosis. Infect Dis lus sanguineus as a potential biologic vector of Ehrlichia platys. Am
Clin North Am. 2008;22:433-448:viii. J Vet Res. 1991;52:1537-1541.
23. Egenvall A, Lilliehook I, Bjoersdorff A, et al. Detection of granulo- 42. Parola P, Cornet JP, Sanogo YO, et al. Detection of Ehrlichia spp.,
cytic Ehrlichia species DNA by PCR in persistently infected dogs. Anaplasma spp., Rickettsia spp., and other eubacteria in ticks
Vet Rec. 2000;146:186-190. from the Thai-Myanmar border and Vietnam. J Clin Microbiol.
24. Heikkila HM, Bondarenko A, Mihalkov A, et  al. Anaplasma 2003;41:1600-1608.
phagocytophilum infection in a domestic cat in Finland: Case 43. Harrus S, Perlman-Avrahami A, Mumcuoglu KY, et al. Molecular
report. Acta Vet Scand. 2010;52:62. detection of Ehrlichia canis, Anaplasma bovis, Anaplasma platys,
25. Tarello W. Microscopic and clinical evidence for Anaplasma Candidatus Midichloria mitochondrii and Babesia canis vogeli in
(Ehrlichia) phagocytophilum infection in Italian cats. Vet Rec. ticks from Israel. Clin Microbiol Infect. 2011;17:459-463.
2005;156:772-774. 44. Chae JS, Yu do H, Shringi S, et al. Microbial pathogens in ticks,
26. Plier ML, Breitschwerdt EB, Hegarty BC, et al. Lack of evidence for rodents and a shrew in northern Gyeonggi-do near the DMZ,
perinatal transmission of canine granulocytic anaplasmosis from a Korea. J Vet Sci. 2008;9:285-293.
bitch to her offspring. J Am Anim Hosp Assoc. 2009;45:232-238. 45. Harrus S, Aroch I, Lavy E, et al. Clinical manifestations of infec-
27. Breitschwerdt EB, Hegarty BC, Hancock SI. Sequential evaluation tious canine cyclic thrombocytopenia. Vet Rec. 1997;141:247-250.
of dogs naturally infected with Ehrlichia canis, Ehrlichia chaffeen- 46. Glaze MB, Gaunt SD. Uveitis associated with Ehrlichia platys
sis, Ehrlichia equi, Ehrlichia ewingii, or Bartonella vinsonii. J Clin infection in a dog. J Am Vet Med Assoc. 1986;189:916-917.
Microbiol. 1998;36:2645-2651. 47. Harvey JW. Anaplasma platys infection. In: Greene CE, ed. Infec-
28. Aguero-Rosenfeld ME. Diagnosis of human granulocytic ehrlichio- tious diseases of the dog and cat. 4th ed. St. Louis, MO: Elsevier
sis: state of the art. Vector Borne Zoonotic Dis. 2002;2:233-239. Saunders; 2012:256-258.
29. Dumler JS, Barat NC, Barat CE, et  al. Human granulocytic 48. Eddlestone SM, Gaunt SD, Neer TM, et al. PCR detection of Ana-
anaplasmosis and macrophage activation. Clin Infect Dis. plasma platys in blood and tissue of dogs during acute phase of experi-
2007;45:199-204. mental infection. Exp Parasitol. 2007;115:205-210.
30. Lepidi H, Bunnell JE, Martin ME, et  al. Comparative pathol- 49. Gaunt S, Beall M, Stillman B, et al. Experimental infection and co-
ogy, and immunohistology associated with clinical illness after infection of dogs with Anaplasma platys and Ehrlichia canis: hemato-
Ehrlichia phagocytophila-group infections. Am J Trop Med Hyg. logic, serologic and molecular findings. Parasit Vectors. 2010;3:33.
2000;62:29-37. 50. Baker DC, Simpson M, Gaunt SD, et  al. Acute Ehrlichia platys
31. Akkoyunlu M, Fikrig E. Gamma interferon dominates the murine infection in the dog. Vet Pathol. 1987;24:449-453.
cytokine response to the agent of human granulocytic ehrlichiosis 51. Chang WL, Su WL, Pan MJ. Two-step PCR in the evaluation of
and helps to control the degree of early rickettsemia. Infect Immun. antibiotic treatment for Ehrlichia platys infection. J Vet Med Sci.
2000;68:1827-1833. 1997;59:849-851.
32. Birkner K, Steiner B, Rinkler C, et  al. The elimination of Ana-
plasma phagocytophilum requires CD4+ T cells, but is independent
of Th1 cytokines and a wide spectrum of effector mechanisms. Eur
J Immunol. 2008;38:3395-3410.
CHAPTER 30

Rocky Mountain Spotted Fever


Linda Kidd and Edward B. Breitschwerdt

terms “rickettsial disease,” “rickettsioses,” and even the term


Overview of Rocky Mountain Spotted Fever “Rickettsia” have been used to refer to several obligately intra-
First Described: R. rickettsii was first described in Montana, cellular organisms including Rickettsia, Bartonella, Ehrlichia,
United States, 19091 (by Ricketts) Anaplasma, Coxiella, and Neorickettsia. Previously, all of these
Cause: Rickettsia rickettsii, a gram-negative, obligately intra- organisms belonged to the order Rickettsiales and most were
cellular bacteria in the family Rickettsiaceae, based on their fastidious or intra-
cellular nature and other characteristics. Therefore, collectively
Affected Hosts: Humans and dogs
members of these diverse genera were referred to as “rickett-
Geographic Distribution: North, Central, and South America sial organisms.”4,5 Recent advances in molecular biologic tech-
Primary Mode of Transmission: Ticks (Dermacentor spp., niques have resulted in the reclassification of several of these
­Rhipicephalus sanguineus, Amblyomma spp.). Most affected organisms. Many have been moved out of the family Rickett-
dogs lack a history of a tick bite. siaceae, and others have been moved out of the order Rickett-
siales. Now the order Rickettsiales includes only two families,
Major Clinical Signs: Rocky Mountain spotted fever (RMSF)
the family Anaplasmataceae, which contains the genera Ana-
is an acute, generally febrile illness. Clinical signs are con-
plasma, Ehrlichia, Wolbachia, and Neorickettsia; and the fam-
sistent with a vasculitis because R. rickettsii infects endo-
ily Rickettsieaceae, which includes the genera Rickettsia and
thelial cells. Major clinical signs include fever, vomiting,
Orientia.2,3,5 Currently, “rickettsial” refers to diseases caused
ocular signs, lymphadenomegaly, splenomegaly, periph-
by organisms in the genera Anaplasma, Ehrlichia, Rickettsia,
eral edema, cutaneous hyperemia and necrosis, polyar-
Neorickettsia, and Orientia, “rickettsioses” refers to diseases
thritis, and neurologic signs. Signs may be complicated by
caused by organisms in the family Rickettsiaceae (Rickettsia
simultaneous infection with other tick-borne pathogens.
and Orientia), and the term “Rickettsia” refers specifically to
Differential Diagnoses: Infection with other tick-borne agents members of the genus Rickettsia.3-5
such as A. phagocytophilum, Ehrlichia spp., ­Bartonella spp., The genus Rickettsia is currently divided into the spotted
Babesia spp., and Borrelia burgdorferi should be con- fever group (SFG) and the typhus group based on phenotypic
sidered. Other causes of severe systemic decompensa- and, more recently, genotypic characteristics.3,5 Some of the
tion and vasculitis, such as sepsis and SIRS due to other phenotypic characteristics that have historically been used to
causes may mimic RMSF. Leptospirosis and other causes group the organisms include the types of vectors that transmit
of vasculitis and thrombocytopenia may also mimic RMSF. them and the pathophysiologic manifestations of the disease.3
Other differential diagnoses for the neurologic signs seen The SFG rickettsial species are transmitted by arthropod (pri-
in RMSF must also be considered. Appropriate antimicro- marily tick) vectors and infect endothelial cells in mammalian
bial treatment with doxycycline must begin before the hosts.5 The two most pathogenic and well-studied SFG rickett-
diagnosis is confirmed by laboratory testing. Misdiagnosis siae are Rickettsia rickettsii, the cause of RMSF in the Western
and delayed or inappropriate antimicrobial drug therapy Hemisphere, and Rickettsia conorii, the cause of Mediterranean
increase morbidity and mortality. spotted fever (MSF) in other areas of the world. The first case of
Human Health Significance: Owners and their physicians RMSF was described in the late 1800s and the first case of MSF
should be contacted whenever RMSF is diagnosed in any was described in the 1920s.6 Therefore, these organisms and
canine patient, because illness in dogs can coincide with their associated diseases are the most well characterized among
or precede illness in humans. the SFG rickettsiae.3 Currently, there are 20 or more species
of SFG rickettsiae.3,6 Some species appear to be nonpathogenic
endosymbionts of ticks.6 However, some SFG rickettsiae previ-
ously thought to be nonpathogenic, such as R. parkerii and R.
Etiologic Agent massiliae, have recently been associated with disease in people.7-9
Previously thought to have rather limited geographic boundar-
Rocky Mountain spotted fever (RMSF) is caused by Rickett- ies, many SFG rickettsiae have also been detected in expand-
sia rickettsii, an obligately intracellular bacteria in the alpha- ing geographic locales around the world.6 For example, RMSF,
proteobacteria (genus Rickettsia, family Rickettsiaceae, order originally described in the Bitterroot Valley of Montana, was
Rickettsiales).2 The terminology used to describe rickettsial dis- subsequently found to be a frequent tick-transmitted infection
ease is confusing and inconsistent due to multiple changes in in the eastern United States, but only recently has transmission
the taxonomic classification of organisms in recent years.3 The via the brown dog tick been documented in the southwestern

300
CHAPTER 30  Rocky Mountain Spotted Fever 301

United States.10,11 Increasing travel and the effects of climate


change on tick populations and habitat are thought to be in part
responsible for this phenomenon.12,13

Epidemiology
Hosts, Life Cycle, and Transmission
Pacific coast tick
Dogs are sentinels for SFG rickettsioses in people. Both (Dermacentor occidentalis)

R. rickettsii and R. conorii infect and cause disease in dogs.14-20


Rickettsia rickettsii infection in dogs can occur before, or coin-
American dog tick
cide with, outbreaks of RMSF in people in the same household (Dermacentor variabilis)
or community.21-24 Several serosurveys in endemic areas have
shown an increased risk of MSF in people who live near dogs
that are seropositive for SFG rickettsiae.25-27 Other SFG rickett-
sial species likely infect dogs, but the extent to which they cause
clinical disease has not yet been established. Species implicated
in natural infection in dogs include R. massiliae, R. japonica,
and R. australis.28-31
Young and purebred dogs are overrepresented in some but
not other studies.17,18 Although some studies suggest male dogs
are at increased risk, no sex predilection has been definitively
documented.17,18,32 Severe disease has been reported in English
springer spaniels with phosphofructokinase (PFK) deficiency
and German shepherd dogs.18,32 Although antibodies to SFG
rickettsiae can be detected in cats that live in endemic areas, A
the ability of SFG rickettsiae to actively infect and cause disease
in cats has not been well characterized.33 Similarly, the abil-
ity of typhus group Rickettsia to cause disease both in dogs
and cats has also not been well characterized and will not be
discussed here.
Rickettsia rickettsii is transmitted by several hard (ixodid)
ticks including Dermacentor variabilis (the American dog tick)
(Figure 30-1, A), Dermacentor andersoni (the Rocky Mountain
Rocky Mountain wood tick
wood tick) (Figure 30-1, B), Amblyomma americanum, Ambly- (Dermacentor andersoni)
omma cajennense, Amblyomma aureolatum, and Rhipicephalus
sanguineus.34-38 Amblyomma cajennense and to a lesser extent,
A. aureolatum transmit the rickettsia in South America. Ticks
that transmit R. rickettsii feed once during each life stage (Figure
30-2). Both transstadial and transovarial transmission occurs in
Dermacentor spp. infected with R. rickettsii, so these ticks serve
as a reservoir of infection.39,40 Dermacentor spp. and Ambly-
omma spp. are three-host ticks. Infection is transmitted among
mammalian hosts and ticks when the tick feeds on different hosts
during each stage of molting. The sylvatic cycle for Dermacentor
ticks involves small mammals, such as chipmunks, pine voles,
mice, and ground squirrels. After organisms are ingested by the
tick, SFG rickettsiae initially replicate in the epithelial cells of
B
the tick midgut, enter the hemolymph and hemocytes, and then
FIGURE 30-1  A, Distribution of Dermacentor variabilis and Dermacentor occidenta-
multiply in tissues.40 Once in the salivary glands, the organism
lis ticks in the United States. B, Distribution of Dermacentor andersoni ticks in the United
is transmitted to a naïve mammalian host on feeding. Transmis- States.
sion can occur within hours of attachment. However, this may
be prolonged (up to 48 hours) when changes in virulence occur
under conditions such as starvation of the tick. These “dor- The role of dogs as a natural reservoir of infection for
mant” rickettsiae undergo a process called reactivation after the R. rickettsii is also unknown.39 Dogs are thought to act as inci-
tick begins feeding.41 Therefore, rapid tick removal can decrease dental hosts when Dermacentor ticks are the vectors, because
the risk of transmission in many, but not all, circumstances. ticks rarely acquire the organism from rickettsemic dogs.43 In
Rhipicephalus sanguineus is a one-host tick, with dogs being contrast, all stages of Rh. sanguineus acquired infection at a high
the preferred host.42 Rh. sanguineus can adapt to hot environ- rate from experimentally infected dogs in one study, but this
ments and commonly resides in walls of housing structures in may have been related to the virulence of the rickettsial strain
close proximity to humans.42 It occasionally feeds on humans, used.38 A relatively high infection rate with SFG rickettsiae in
especially when ambient temperatures are high.12 The role of this naturally infected Rh. sanguineus has also been described.37,44
tick as a reservoir for R. rickettsii in nature has yet to be elucidated. Therefore, dogs may play a role in maintenance of a reservoir
302 SECTION 2  Bacterial Diseases

Adults attach to
7
third host for feeding
and mating

Nymphs molt
into adults
6 1 Adult females
drop off host
to lay eggs

5 2 Eggs hatch
Nymphs feed into larvae
on second host
and may acquire
R. rickettsii 3 Larvae feed
4 on first host
Larvae molt and may acquire
into nymphs R. rickettsii
FIGURE 30-2  Transmission cycle of Rickettsia rickettsii in tick populations, people, and dogs. After adult female ticks lay eggs and they hatch (1 and 2), Dermacentor spp. ticks acquire
R. rickettsii upon feeding as larvae or nymphs on rodents (3); they then transmit during subsequent feeding as nymphs or adults (4 and 6). Adult ticks feed on a variety of canid species, such as
foxes and possibly wolves and coyotes. In addition, R. rickettsii infection can be maintained transovarially in Dermacentor ticks, so some larvae that hatch from the egg mass already harbor
an infection. (Redrawn from Nicholson WL, Allen KE, McQuiston JH, et al. The increasing recognition of rickettsial pathogens in dogs and people. Trends Parasitol 2010;26[4]:205-212.)

Alaska

0 cases
1 - 100 cases
Hawaii
100 - 500 cases
500 - 1000 cases
> 1000 cases
FIGURE 30-3  Geographic distribution of the incidence of spotted fever group rickettsioses in people in the United States, 2005-2009. (Compiled from data from the Centers for Disease
Control and Prevention, Morbidity and Mortality Weekly Reports.)

of R. rickettsii infection in endemic foci where Rh. sanguineus importance of D. variabilis as vectors in endemic areas has
is the primary vector. recently been questioned.45 Although disease can occur any time
Direct transmission of R. rickettsii to people has been of year, most cases of canine and human RMSF are reported
described in laboratory settings through aerosolization. Direct from April through October, months of peak tick activity.17,46,47
transmission from blood or other contaminated biologic prod- Dogs that live outdoors, particularly those with access to shrubs
ucts also has the potential to occur. and high grass, are at increased risk.17
In recent years the incidence of RMSF in people has
Incidence and Geographic Distribution increased.47 Some of the increase may be due to misdiagnosis
Rocky Mountain spotted fever occurs in North, Central, and of disease caused by species of SFG rickettsiae other than R.
South America (where it is known as Brazilian spotted fever). ­rickettsii.48,49 A similar increase in seroprevalence rates has been
In North America, most cases of RMSF occur in the southeast- documented in dogs suspected to have a tick-borne infection in
ern and south central states (Figure 30-3). Disease distribution the United States.50 The geographic distribution of RMSF in
in the United States primarily follows the distribution of the the United States has also increased beyond the distribution
primary vectors, D. variabilis and D. andersoni, although the of D. variabilis and D. andersoni. Rhipicephalus sanguineus
CHAPTER 30  Rocky Mountain Spotted Fever 303

is the main vector for R. rickettsii in some parts of the West- important in maintaining vascular integrity and energy metabo-
ern Hemisphere, including Mexico and now the south- lism in endothelial cells under hypoxic or oxidative stress,60,61
western United States. In South America, Amblyomma which may explain the predisposition of English springer span-
species are the main tick vector. Tick species other than iels with PFK deficiency to severe disease. Vasculitis associated
D. variabilis, D. ­andersoni, and Rh. sanguineus are also with R. rickettsii infection manifests as disordered primary
infected with R. ­rickettsii in the United States.44 For example, hemostasis, tissue edema, hypovolemia, and microthrombosis.
A. ­americanum transmitted RMSF to a person in North Car- Increased vascular permeability and the associated edema
olina.36 An outbreak in Mexicali, Mexico, was attributed to and hypovolemia results from disruption of adherens junc-
R. rickettsii transmitted by Rh. sanguineus ticks associated tions, endothelial cell death, expression of inflammatory cyto-
with dogs.51 This prompted ongoing surveillance efforts in kines such as Il1-β, IFN-γ, and TNF-α, and induction of COX-2
ticks, dogs, and people in southern California, a nonendemic with subsequent prostaglandin production.55 Microthrombosis
area for RMSF. Thus far, no R. ­rickettsii has been found in results from altered platelet adherence to endothelium, increased
Rh. ­sanguineus ticks from dogs residing in animal shelters tissue factor expression, increased plasminogen activator inhibi-
located just across the U.S. border from the outbreak.52,53 This tor, and the release of von Willebrand’s factor.55
may be due to subtle differences in microenvironment or other Low numbers of organisms circulate in blood for approxi-
factors that affect regional infection rates in ticks.54 mately 13 days after infection, which includes the time that
clinical signs are observed.19,62,63 Organisms are free and also
Clinical Features contained within circulating endothelial cells, which are thought
to be released from the vessels because of decreased adhesion
Signs and Their Pathogenesis after rickettsial invasion.57 Thus, RMSF is an acute disease.
The endotheliotropism associated with these bacteria results in Chronic infection has not been documented in dogs or people.
the characteristic clinical sign of infection, which is disseminated Co-infection with other vector-borne agents is common and
vasculitis. In people, cutaneous macules, papules, and petechia- should be considered if the clinical signs are atypical, if there is an
tion that occur in association with vasculitis form a rash that incomplete response to doxycycline therapy, or if clinical signs
looks like “spots,” hence the name Rocky Mountain spotted have been present for a week before the time of evaluation.17,64
fever.6 The clinical signs, time course of illness, and response to Because of variation in the extent and severity of vascular
therapy for RMSF in dogs are very similar, if not identical, to injury among dogs, a range of signs can occur, and importantly,
those associated with RMSF in people.17-20,34 However, cutane- disease manifestations are initially mild and nonspecific.15,17,18
ous lesions are not always present in dogs or people (“Rocky Often, there is no known history of a tick bite. Therefore, the
Mountain spotless fever”). In people, spotted fever rickettsioses clinician (physician and veterinarian) must have a high index of
caused by organisms other than R. rickettsii are often associ- suspicion in order to correctly diagnose and treat this disease.
ated with eschar (an area of cutaneous necrosis) formation.2,13 This is very important because a delay in diagnosis and appropri-
Clinicians should keep in mind that dogs from nonendemic ate antimicrobial therapy dramatically increases morbidity and
areas that have classic signs of RMSF, or those that present with mortality in people and in dogs.17,46 A “One Health” approach
atypical clinical signs, may be infected with R. rickettsii or other to the management of canine and human RMSF is clearly logical.
SFG rickettsial species. Lethargy and anorexia are common and may be the only clinical
Once the organism is inoculated, it spreads through lym- signs. Vomiting and diarrhea occur frequently in dogs and people
phatics or directly into the bloodstream to the small capillaries. with RMSF. Melena may be observed, as may a variety of CNS
R. rickettsii primarily infects endothelial cells, although smooth abnormalities including vestibular disease and seizures.17,18,20
muscles and monocytes may also be infected.2,55 The bacterial Dramatic and rapid weight loss has been described.20
outer membrane protein A (OmpA) and outer membrane pro-
tein B (OmpB) are important for attachment, adhesion, and vir- Physical Examination Findings
ulence.56,57 These proteins are also responsible for differences in Fever is present in approximately 80% of naturally infected
serotype, and antibodies to OmpB confer immunity to infection dogs. Ocular signs are also frequently observed and may
in experimental settings. SFG rickettsiae enter cells by induc- include a mucopurulent discharge, scleral and conjunctival
ing phagocytosis and are released into the cytoplasm through injection and hemorrhage, conjunctivitis, uveitis, retinal hem-
the action of enzymes such as phospholipase D and hemolysin orrhage, and retinitis (Figure 30-4). Lymphadenomegaly and
C.57 The bacteria live in the cytoplasm and the nucleus, deriving splenomegaly also occur. Respiratory abnormalities include
nutrients and energy from the host cell.2,56 They spread from nasal discharge, epistaxis, and tachypnea. Mucocutaneous and
cell to cell by inducing actin to polymerize, which pushes the cutaneous abnormalities include petechiae, ecchymosis, periph-
bacteria directly into adjacent cells.57,58 This helps them evade eral edema (which can be localized over a joint, the prepuce,
the immune response and to disseminate without rupturing the or the mammary chain), hyperemia, and necrosis. Gangrenous
cell. They are also released into circulation when they exit the necrosis can be so severe as to require reconstructive surgery
luminal surface of the cell membrane or when endothelial cell after successful treatment of the acute febrile illness.15-18,20,65,66
death or detachment occurs.57 Spotted fever group rickettsiae Orchitis and scrotal edema, hyperemia, and epididymal pain are
activate the transcription factor NFκB, which inhibits apoptosis common in intact male dogs and should prompt consideration
and fosters further growth of the organism.56,57 of RMSF when present. Generalized myalgia and arthralgia can
Damage to endothelial cells leads to vasculitis and an increase be observed. Arrhythmias may also be detected. CNS abnor-
in microvascular permeability. Mechanisms of cellular damage malities can be focal or generalized and include paraparesis,
include oxidative injury through the production of reactive tetraparesis, ataxia, hyperesthesia, ataxia, central or peripheral
oxygen species, cellular necrosis, and induction of endothelial vestibular signs, stupor, seizures, and/or coma. Neurologic signs
apoptosis by CD8+ T cells.55-57,59 Activation of PFK may be are more common in dogs with a high R. rickettsii antibody
304 SECTION 2  Bacterial Diseases

titer, which suggests a longer duration of illness or a delay in high index of suspicion based on clinical signs is necessary, because
diagnosis.17 Residual neurologic deficits may occur after infec- treatment must be instituted before the results of diagnostic tests
tion in severely affected individuals. Microvascular hemorrhage, (including rapid PCR assays) confirm infection.17,20 Co-infection
thrombosis, hypotension, oliguric renal failure, cardiovascular with other vector-borne disease agents should be considered in
collapse, and coma can occur terminally. patients who fail to respond rapidly to treatment (within the first
24 to 48 hours after initiation of doxycycline treatment).64 Because
Diagnosis direct inoculation into blood or aerosolization can cause infection,
all specimens should be handled with care and marked clearly as
A combination of diagnostic testing is often necessary to confirm biohazards. Needle-stick injuries, contact with cuts in the skin, and
infection by SFG rickettsiae. Active infection is confirmed in a aerosolization of rickettsemic blood should be avoided.
patient with compatible acute clinical signs and demonstration of
the organism using PCR assays or immunohistochemistry (IHC), Laboratory Abnormalities
or documentation of seroconversion (Figure 30-5). Importantly, a Complete Blood Count
Thrombocytopenia is common in RMSF and occurs due to vas-
culitis and immune-mediated platelet destruction.67 However,
it does not occur in all dogs with RMSF.17,20 The white blood
cell count may initially decrease and then tends to increase with
duration of illness.18-20 Neutrophils may have toxic change.17
Despite the acute nature of this severe illness, a nonregenera-
tive anemia may be present and persist until the dog is treated
appropriately and begins to recover.15,17,20

Serum Biochemical Tests


Serum biochemical abnormalities can include hypoalbuminemia
(due to vasculitis or protein-losing nephropathy), increased ALP
A activity, hyponatremia, and mild hyperbilirubinemia.15,17,20,65
Hyponatremia has been associated with the syndrome of inap-
propriate antidiuretic hormone secretion (SIADH) in people
with RMSF.

Urinalysis
Urinalysis results in dogs with RMSF are variable and may
include proteinuria, hematuria and bilirubinuria, and pyuria.
Granular casts can be observed.17,18

Coagulation Profile
Coagulation abnormalities include prolonged APTT and
increased serum fibrinogen concentration. Less commonly, pro-
longed PT and increased fibrin degradation products are observed.
Although a prothrombotic state can occur during fulminant dis-
ease, disseminated intravascular coagulation is uncommon.15,17

Body Fluid Cytology


B Cytologic examination of aspirates from enlarged lymph
FIGURE 30-4  Ocular complications in dogs with RMSF. A, Conjunctival hyperemia nodes in dogs with RMSF is consistent with reactive lym-
and scleral injection. B, Retinal hemorrhages. phoid hyperplasia.17,18 Arthrocentesis may reveal neutrophilic

Clinical Signs Convalescence


DPI 0 23 17 36+
Culture

PCR*

Tissue IFA

Serology

Tick bite!
FIGURE 30-5  Timing in relation to day postinfection (DPI) that clinical signs and diagnostic tests for Rickettsia rickettsii may be positive in an infected dog16,62,63,70,76
Further studies are needed to determine exactly how DNA can be detected in peripheral blood after infection.62,63
CHAPTER 30  Rocky Mountain Spotted Fever 305

polyarthritis.17,18 Cerebrospinal fluid analysis in dogs with neu- seroconversion in dogs infected with R. rickettsii.19 Because
rologic signs may reveal a mixed cellular, neutrophilic, or lym- of a lack of specificity, assays that detect IgM alone cannot be
phocytic pleocytosis.17,18,20,68 used to accurately diagnose acute RMSF.70 There is extensive
serologic cross-reactivity among SFG rickettsiae, and thus
Diagnostic Imaging a positive titer only indicates exposure to a SFG rickettsial
Thoracic radiographs in dogs with RMSF may show an unstruc- species. In general, the infecting species is presumed based
tured interstitial pattern.18,69 Testicular ultrasound findings may on geographic locale, so RMSF is the presumptive diagnosis
be consistent with orchitis in intact males.66 in dogs that seroconvert in the southeastern United States.
However, other species of SFG rickettsiae may also be pres-
Microbiologic Tests ent and induce disease.28,71 Exposure to nonpathogenic SFG
Diagnostic assays available for RMSF in dogs are shown in rickettsiae, some of which are common endosymbionts in
Table 30-1. Because of the pathogenesis of the infection, labo- ticks, may be a common cause of positive titers, particularly
ratory diagnostics (including serology and PCR assays) may not low and persistent titers, to R. rickettsii in healthy people
indicate infection is present at the time that clinical signs mani- and dogs.49,72 Serologic cross-reactivity with SFG rickettsiae
fest (see Figure 30-5). In addition, most diagnostic tests do not also may occur in dogs and people infected with Bartonella
differentiate among species of SFG rickettsiae. henselae.73 In addition, previous infection with R. rickettsii
may result in persistent antibody titers.70 Thus, infection
Culture with R. rickettsii cannot be definitively diagnosed based on
Because of the obligately intracellular and extremely pathogenic a single positive titer. However, a single high titer in the con-
nature of R. rickettsii, culture is difficult to perform and process- text of acute and compatible clinical signs and an appropriate
ing of specimens for isolation is limited to BSL 3 facilities (see response to therapy suggests infection.17 Titers are commonly
Table 1-4). Therefore, culture is not commonly used for routine negative early in the course of infection, because clinical signs
diagnosis. Currently, R. rickettsii is listed as a Category A Bio- often occur before seroconversion in naturally and experi-
terrorism agent, so all isolates must be reported to the Centers mentally infected dogs.20,70,74 Thus, acute and convalescent
for Disease Control and Prevention or destroyed immediately. serology (run in the same laboratory and ideally in the same
batch) and documentation of a fourfold change in titer is nec-
Serologic Diagnosis essary to confirm acute SFG rickettsiae infection. The conva-
Indirect microimmunofluorescence (MIF) assays that detect lescent serum specimen should be drawn 2 to 3 weeks after
IgM and IgG antibodies are most commonly used to document the acute specimen.

TABLE 30-1
Diagnostic Assays Available for Rocky Mountain Spotted Fever in Dogs
Assay Specimen Type Target Performance
Indirect Serum IgM and IgG a­ ntibodies Demonstration of a fourfold change in titer (seroconversion)
microimmuno­ against R. rickettsii is very sensitive. False-negative results are common during
fluorescence the acute phase of infection. A positive antibody response
assay (MIF) may reflect prior exposure to a nonpathogenic SFG rick-
ettsia, or long-lived circulating antibody from previous
infection with SFG rickettsiae. Cross-reactivity to other
SFG rickettsiae and to B. henselae occurs.
PCR Whole blood Gene target varies In vitro sensitivity may approach 100%; however, absolute
with testing (clinical) sensitivity is lower, approximately 60%, because
laboratories organisms circulate in blood in low numbers and tran-
siently during the acute phase of infection. Sensitivity is fur-
ther reduced by antimicrobial drug treatment. Specificity
approaches 100%, particularly if the laboratory sequences
all PCR amplicons. PCR testing for other tick-borne
organisms that cause similar clinical signs is available as
PCR panels from some testing laboratories. Only a few
PCR assays differentiate among Rickettsia species. Nega-
tive PCR assay results do not rule out infection.
Histopathology Cutaneous biopsy Characteristic Direct IFA sensitivity is approximately 80% during acute
with direct IFA (inguinal or flank ­perivascular infection. Biopsy of a petechial or ecchymotic lesion may
or Gimenez targeting areas ­inflammation and increase sensitivity. False negatives occur if organisms are
­staining with lesions) ­vasculitis with absent from the lesion because of random chance, timing
or necropsy ­necrosis and p
­ resence of specimen collection, or prior antimicrobial treatment.
­specimens of the organism Does not differentiate among species of SFG Rickettsia.
306 SECTION 2  Bacterial Diseases

Molecular Diagnosis Using Polymerase Chain Reaction Treatment


PCR testing of whole blood can be used to confirm infection
in seronegative dogs during the acute phase of disease.62,63,74 Antimicrobial Treatment
A sensitive PCR assay that detects and differentiates among Appropriate antibiotic therapy must be immediately instituted
SFG rickettsiae that infect dog blood has been described, and a based on clinical suspicion, and before diagnostic tests con-
variety of PCR assays are available through commercial veteri- firm infection. Inappropriate or delayed antibiotic therapy may
nary diagnostic laboratories.74 However, because R. ­rickettsii increase morbidity and mortality.17,46 Some antimicrobial drugs
circulates in blood in low numbers and only transiently, the such as trimethoprim sulfonamides may actually worsen dis-
sensitivity of any diagnostic assay to detect infection in the ease progression in human patients.46 Doxycycline is the treat-
bloodstream is limited. One real-time PCR assay, which could ment of choice (Table 30-2). It effectively eliminates infection
detect as few as five copies of organism DNA, had a sensitiv- and is active against A. phagocytophilum, Ehrlichia spp., and
ity of only 53% for detection of R. rickettsii in acute samples B. ­burgdorferi, which may be present in co-infections or cause
from naturally infected dogs with signs of RMSF.74 The sen- disease that resembles RMSF. Seven days of treatment is ade-
sitivity of another R. rickettsii PCR assay for detection of quate in most cases. Treatment a few days past defervescence is
R. rickettsii in experimentally infected dogs ranged from 33% to recommended. A longer course of treatment is recommended for
100% depending on the day of sampling.63 Furthermore, false dogs that are co-infected with Ehrlichia spp. or B. ­burgdorferi.
negatives can occur after initiation of appropriate antimicrobial Chloramphenicol was effective in experimentally infected
treatment. Therefore, although a positive PCR assay result con- dogs.76 However, it may be less effective than doxycycline for
firms infection, a negative result does not exclude the diagnosis. treating RMSF in people and has less activity in  vitro against
Some PCR assays can differentiate among infecting Rickettsia Ehrlichia chaffeensis and A. phagocytophilum.46 Enrofloxacin
species, whereas others only amplify conserved rickettsial DNA was effective for treatment of RMSF in experimentally infected
targets.62,74 Clinicians should consult with laboratories to deter- dogs.76 However, enrofloxacin is not effective for treatment of
mine the in  vitro (analytical) and in  vivo (clinical) sensitivity E. canis infections.77 Use of parenteral antimicrobial drugs may
of a PCR assay and its ability to differentiate among infecting be necessary in severely debilitated or vomiting patients.
Rickettsia species.
Supportive Care
Pathologic Findings Many dogs require hospitalization.20 Aggressive supportive care
Gross Pathologic Findings for complications such as thrombosis, CNS deficits, and gastro-
Gross pathologic findings in dogs with RMSF are consistent intestinal signs may be necessary. Because of the loss of vascular
with vasculitis and may include edema, particularly of the integrity, fluids should be administered with caution, and colloids
ears, muzzle, and scrotum, and ecchymosis, petechiae, and/or may be warranted in some cases. The clinician should avoid exac-
focal hemorrhages of the skin, mucous membranes, and viscera erbation of interstitial edema, which can contribute to cerebral
(including the brain). Lymphadenomegaly and splenomegaly edema and death. The use of glucocorticoids in dogs with RMSF
are also common findings.15,16,20,22 is controversial. Antiinflammatory and immunosuppressive doses
did not affect overall outcome experimentally infected dogs, but
Histopathologic Findings rickettsemia was prolonged in dogs concurrently treated with
Microscopically, the predominant lesion is vasculitis. Neutro- immunosuppressive doses of glucocorticoids and doxycycline.78
phils, monocytes, and/or lymphoreticular cells predominate in Antiinflammatory doses of glucocorticoids have been used in
the inflammatory lesions. The inflammation may surround and dogs with severe CNS manifestations and may be necessary topi-
invade large small and medium-size vessels of multiple organs cally or systemically for treatment of ocular abnormalities.68,79
and tissues and is frequently accompanied by focal necrosis
and hemorrhage. Meningoencephalitis, splenitis, myocardial Prognosis
necrosis, glomerulonephritis, and renal vasculitis have been The response to appropriate antibiotic therapy in dogs
described.15,20,22 with RMSF is rapid (24 to 48 hours). Co-infection with
Direct IFA can be used to detect organisms in skin and other
organs. Identification of abundant SFG rickettsiae within and
around small to medium-size vessels and vascular endothelial TABLE 30-2
cells can confirm infection.20 In experimentally infected dogs,
the sensitivity of IHC was 78.3%, and organisms could be
Antimicrobials Used to Treat Rocky Mountain Spotted
visualized between days 7 and 12 of infection (days 3 to 8 of Fever in Dogs
fever).75 In that study, specimens collected from areas other
than the central focus of vasculitis were negative. However, in Dose Interval Duration
naturally infected dogs, the sensitivity was 80% for skin speci- Drug (mg/kg) Route (hours) (days)
mens from the inguinal region. Half of those specimens lacked Doxycycline 5 PO, IV 12 7 to 14
gross lesions.18 Thus, the diagnostic sensitivity of IHC is most Chloramphenicol* 30 PO, IV 8 7 to 14
likely enhanced by obtaining lesional biopsies, but may not be
decreased when a lesion is not biopsied. The sensitivity of direct
Enrofloxacin 3† to 5 PO, IV 12 7 to 14
IHC is decreased by antibiotic therapy. IHC does not differenti-
ate between SFG rickettsial species. Gimenez stain can also be *May cause aplastic anemia in humans (wear gloves); may be less
used to visualize rickettsial organisms in tissues or tissue culture ­effective than doxycycline.
isolation attempts, but this stain is not rickettsia specific and †Dose used in experimentally infected dogs.76 Caution required in young

does not allow for differentiation among species. animals (see Chapter 8).
CHAPTER 30  Rocky Mountain Spotted Fever 307

B. burgdorferi, Ehrlichia spp., B


­ abesia spp., and Bartonella spp. when exposure to R. rickettsii is a possibility. In cases where
should be considered in dogs with severe or prolonged clinical Rh. sanguineus is the vector, environmental control of ticks is
signs or dogs that fail to respond to doxycycline. Residual CNS particularly important.
and other deficits may occur in severely affected patients.17 The
prognosis is good to excellent if the disease is diagnosed and Public Health Aspects
treated with appropriate antibiotics and supportive care early in
the course of illness.17,68 People living near dogs and with dogs in endemic areas are
at increased risk for acquiring RMSF. This is likely due to
Immunity and Vaccination increased human contact with ticks through interaction with
tick-infested dogs. Also, because of transovarial transmis-
Cell-mediated and innate immunity is important in the clearance sion, dogs and people can independently acquire the infec-
of rickettsial infections. CD4+ and CD8+ cells, along with macro- tion from different ticks questing in the same environment.
phages and dendritic cells, are believed to be sources of inflamma- Because dogs have higher exposure to ticks than their human
tory cytokines such as IFN-γ, TNF-α, Il-1β, and RANTES (CCL5). counterparts, they serve as excellent environmental sentinels
These cytokines increase production of nitric oxide synthetase and for RMSF.21,22,24,71 In the United States, the diagnosis of SFG
hydrogen peroxide by the endothelial cell, which helps to elimi- rickettsioses in humans is notifiable to public health authori-
nate the organisms.56,57 The endothelial cells themselves also pro- ties. Some counties in certain states also require reporting
duce cytokines such as IL-6, Il-8, and MCP-1 (CCL2) that recruit for dogs, particularly during suspected outbreaks.81 From a
immune cells and combat infection. Antibodies are not thought public health standpoint, it is important for veterinarians to
to be important initially for clearance of infection because they confirm a diagnosis of RMSF in a dog whenever possible, and
form after the fulminant stages of disease.57 However, antibod- to warn the family of the increased risk of acquiring a R. rick-
ies are long-lived and together with cell-mediated immunity may ettsii–infected tick in the same location as the dog acquired
prevent subsequent infection. Immunity to reinfection is thought the disease. Veterinarians must communicate with owners and
to be lifelong in people, and experimentally infected dogs did not physicians that infection in a dog may precede tick-transmitted
develop illness following rechallenge at 6 months and 3 years infection to owners or neighbors. Clients should be instructed
after the initial infection. Vaccination is not available for dogs or to remove ticks properly. Education of owners with regard to
people currently, but may be in the future.80 the importance of tick control and prevention for both peo-
ple and pets and the environment is critical. Novel and well-
Prevention characterized species of SFG rickettsiae are important causes
of emerging infectious disease in humans. Furthermore, R.
Avoidance of tick-infested areas and routine inspection of dogs rickettsii is considered a potential bioterrorist agent.80 Because
for ticks after outdoor activities can help to prevent RMSF. The dogs are sentinels for infection, veterinarians play an impor-
reader is referred to Chapter 28 for information on tick preven- tant role in detecting, defining, and preventing illness in their
tion and safe tick removal. Safe tick removal is especially critical canine patients and their human companions.

CASE EXAMPLES pain score of 1 out of 4, rectal temperature of 103.7°F


(39.8°C), HR 130 beats/min, tachypnea (42 breaths/min),
Rocky Mountain Spotted Fever in a Dog and a normal CRT. The body condition score was 5/9.
The dog stood in a hunched position and had a stiff gait,
Signalment: A 5-year-old MC Australian shepherd from North but localizing pain was not elicited on palpation of the
Carolina spine, joints, or long bones. There was no nasal or ocular
History: The dog was examined by the North Carolina State discharge, no petechiae or rash, and no obvious swelling
University veterinary emergency service on April 11 because or joint effusion. Popliteal lymph nodes were prominent,
of recent onset of lethargy, fever, tachypnea, and a stiff gait but there were no other abnormalities noted on physical
when walking. Historically the dog had been healthy, had examination. Systolic blood pressure was 153 mm Hg, and
received routine vaccinations, heartworm preventive, and a diastolic and mean blood pressures were 93 mm Hg and
flea acaricide, and ran approximately 3 miles each evening 111 mm Hg, respectively.
with the owner, who was a veterinarian. On the day of Laboratory Findings: The hematocrit was 42%, total
presentation and several hours after the evening run, the plasma protein was 6.2 mg/dL, and the platelet count was
dog became lethargic, refused to eat dog food or treats, and 120,000/µL. A serum biochemical profile showed only
seemed painful upon manipulation. The owner noticed that hypoglobulinemia (1.7 g/dL).
the dog’s heart rate, which was normally 50 to 60 beats/min, Microbiologic Testing: A SNAP 4Dx (IDEXX Laboratories)
was 120 beats/min at rest. Rectal temperature at home just in-house ELISA assay was negative. Indirect IFA assays for
prior to presentation was 104.4°F (40.2°C). Three days before Babesia canis, Bartonella henselae, Bartonella vinsonii subsp.
the dog was evaluated, the owner had removed a non- berkhoffii, and Ehrlichia canis were negative (no detectable
engorged tick from the dog. antibodies at a 1:16 screening dilution). The dog was
Physical Examination: Physical examination findings seroreactive to Rickettsia rickettsii antigens at a titer of 1:64.
included a body weight of 21.3 kg, 3%-5% dehydration, A PCR panel capable of detecting Anaplasma spp., Babesia
308 SECTION 2  Bacterial Diseases

spp., Ehrlichia spp., and Rickettsia spp. was negative on bed until the next morning. These symptoms became pro-
whole blood. A convalescent R. rickettsii antibody titer was gressively more severe during the day and the tick attach-
1:256, which supported a diagnosis of SFG rickettsiosis, ment site had developed into an erythematous, circular le-
most likely RMSF. sion with induration and a necrotic center, consistent with a
Treatment: Because of the acute onset of lethargy and fever, rickettsial eschar. Due to the history of tick attachment and
the history of recent tick attachment, the documentation fever (maximum temperature 102.9°F [39.4°C]), a spotted
of thrombocytopenia, and the relatively high frequency of fever rickettsiosis, such as RMSF, or neutrophilic or mono-
RMSF in dogs and people in North Carolina, doxycycline cytic ehrlichiosis, caused by Ehrlichia ewingii and Ehrlichia
(4.8 mg/kg PO q12h) was prescribed for 3 weeks before chaffeensis, respectively, was suspected. Neutropenia (2553
the results of microbiologic testing became available. The cells/µL) was accompanied by a left shift (5% band neutro-
dog responded rapidly after doxycycline treatment, with phils) and thrombocytopenia (148,000/µL). Doxycycline
appetite and behaviors normalizing within 24 to 48 hours was dispensed with instructions to take 100 mg q12h for
after the first dose. 7 days. That evening, after sleeping for 6 hours, the patient
Comment: Although the clinical signs in this dog were ate a small quantity of food, after which he became severely
relatively nonspecific, the clinical presentation was very nauseated and fainted, and impact with the floor was asso-
typical of RMSF. Because R. rickettsii causes generalized ciated with a severe blow to the back of the head. The next
vascular injury resulting in increased vascular permeability, day (May 19) a maculopapular rash appeared that predomi-
fluid and protein leakage out of the intravascular space, and nantly involved the inferior portions of the arms and legs.
an acute neutrophilic inflammatory response throughout Throughout the day the distribution of the rash spread and
tissue sites within the body, pain that appears to shift the severity progressed from barely visible to obvious over
in location (back pain, joint pain, abdominal pain, neck most of the body. At no time did the rash involve the palms
pain) is a typical disease manifestation. Also, the hunched or plantar surface of the feet. Fever resolved within 36 hours
appearance reported in this dog is commonly observed after the initiation of doxycycline, and the rash began to
in dogs with RMSF. As this dog was owned by veterinarian fade gradually after 48 hours of treatment. Within a week
and a tick had been removed a few days earlier, a clinical after starting doxycycline, the patient was experiencing
diagnosis and appropriate treatment were initiated earlier in no symptoms and had no sequelae as a result of the infec-
the course of illness than is typical of most dogs with RMSF. tion or the fall. A repeat CBC (May 25) identified a normal
The rapid initiation of doxycycline may have blunted the neutrophil count (4378 cells/µL), no band neutrophils, and
humoral antibody response and decreased the convalescent a normal platelet count (320,000/µL). Acute-phase serum
antibody titer to a level that is lower than expected in most was collected and stored until the convalescent sample was
cases of RMSF in dogs. It is important to note that PCR assay obtained. Rickettsia and Ehrlichia spp. PCR was performed
is insensitive in the early stages of RMSF, as there are often immediately. PCR amplicons, obtained from the patient’s
inadequate numbers of organisms in the blood to achieve blood and from the tick, were sequenced, confirming the
successful amplification of rickettsial DNA. A very rapid presence of R. rickettsii DNA. PCR for Ehrlichia spp. DNA was
response to doxycycline is expected, unless there has been a negative from both the tick and the patient. Subsequently,
delay in diagnosis, accompanied by the onset of neurologic seroconversion to R. rickettsii antigens was identified (1:64
abnormalities. Dogs that develop neurologic complications acute titer, convalescent titer 1:512 after 4 weeks). The pa-
experience a more prolonged recovery and can have tient did not seroconvert to Ehrlichia spp. antigens.
residual neurologic deficits. Comment: Historically, transmission of R. rickettsii in North
America was attributed solely to Dermacentor variabilis in
Rocky Mountain Spotted Fever in a Human Patient the eastern United States and to Dermacentor andersoni
On May 9, a 61-year-old man, who resided on a farm in Wake in the western United States. However, between 2002 and
County, North Carolina, removed an embedded tick from 2004, researchers at the Centers for Disease Control and
the hairy portion of his right armpit. Although duration of Prevention documented Rh. sanguineus transmission of R.
attachment was unknown, it was likely that the tick was rickettsii to people residing in Arizona. Although this is most
acquired 2 to 3 days earlier while pulling weeds from a hay likely an infrequent occurrence, this patient was infected
field. Using a tick identification key, an experienced research with R. rickettsii by A. americanum. It is important to note
technician classified the tick as a male ­Amblyomma ameri- the similarities between the historical, physical examination
canum and stored the tick in a vial containing alcohol. On and laboratory findings for the dog discussed previously
May 16, while working on the farm, the patient experienced and the human patient. In several published case reports,
mild nausea after drinking water and became transiently dogs develop RMSF before a family member contracts the
dizzy. The next morning chills developed, and by the mid- infection from a tick. Thus, it is important for veterinarians
afternoon the patient became febrile (101.2°F [38.4°C]), to recognize and confirm a diagnosis of RMSF in dogs and to
developed muscle pain, a mild headache and remained in educate the client as to the risk of tick exposure within their
local environment.
CHAPTER 30  Rocky Mountain Spotted Fever 309

SUGGESTED READINGS 19. Breitschwerdt EB, Walker DH, Levy MG, et al. Clinical, hemato-
logic, and humoral immune response in female dogs inoculated
Gasser AM, Birkenheuer AJ, Breitschwerdt EB. Canine Rocky Moun- with ­Rickettsia rickettsii and Rickettsia montana. Am J Vet Res.
tain spotted fever: a retrospective study of 30 cases. J Am Anim Hosp 1988;49(1):70-76.
Assoc. 2001;37(1):41-48. 20. Breitschwerdt EB, Meuten DJ, Walker DH, et  al. Canine Rocky
Elchos BN, Goddard J. Implications of presumptive fatal Rocky Moun- Mountain spotted fever: a kennel epizootic. Am J Vet Res.
tain spotted fever in two dogs and their owner. J Am Vet Med Assoc. 1985;46(10):2124-2128.
2003;223(10):1450-1452:1433. 21. Elchos BN, Goddard J. Implications of presumptive fatal Rocky
Nicholson WL, Allen KE, McQuiston JH, et al. The increasing recogni- Mountain spotted fever in two dogs and their owner. J Am Vet
tion of rickettsial pathogens in dogs and people. Trends Parasitol. Med Assoc. 2003;223(10):1450-1452:1433.
2010;26(4):205-212. 22. Paddock CD, Brenner O, Vaid C, et  al. Short report: concurrent
Rocky Mountain spotted fever in a dog and its owner. Am J Trop
REFERENCES Med Hyg. 2002;66(2):197-199.
1. Ricketts HE. Some aspects of Rocky Mountain spotted fever as 23. Demma LJ, Traeger M, Blau D, et al. Serologic evidence for expo-
shown by recent investigations, 1909. Rev Infect Dis. 1991;13(6): sure to Rickettsia rickettsii in eastern Arizona and recent emergence
1227-1240. of Rocky Mountain spotted fever in this region. Vector Borne Zoo-
2. Bechah Y, Capo C, Mege JL, Raoult D. Rickettsial diseases: from notic Dis. 2006;6(4):423-429.
Rickettsia-arthropod relationships to pathophysiology and animal 24. Nicholson WL, Gordon R, Demma LJ. Spotted fever group rickett-
models. Future Microbiol. 2008;3(2):223-236. sial infection in dogs from eastern Arizona: how long has it been
3. Raoult D, Fournier PE, Eremeeva M, et al. Naming of Rickettsiae there? Ann N Y Acad Sci. 2006;1078:519-522.
and rickettsial diseases. Ann N Y Acad Sci. 2005;1063:1-12. 25. Raoult D, Toga B, Dunan S, et  al. Mediterranean spotted fever
4. Jensenius M, Davis X, von Sonnenburg F, et  al. Multicenter in the South of France; serosurvey of dogs. Trop Geogr Med.
GeoSentinel analysis of rickettsial diseases in international travel- 1985;37(3):258-260.
ers, 1996-2008. Emerg Infect Dis. 2009;15(11):1791-1798. 26. Mannelli A, Mandola ML, Pedri P, et  al. Associations between
5. Fournier PE, Raoult D. Current knowledge on phylogeny and tax- dogs that were serologically positive for Rickettsia conorii relative
onomy of Rickettsia spp. Ann N Y Acad Sci. 2009;1166:1-11. to the residences of two human cases of Mediterranean spotted
6. Parola P, Paddock CD, Raoult D. Tick-borne rickettsioses around fever in Piemonte (Italy). Prev Vet Med. Jul 30 2003;60(1):13-26.
the world: emerging diseases challenging old concepts. Clin Micro- 27. Segura-Porta F, Diestre-Ortin G, Ortuno-Romero A, et  al. Prev-
biol Rev. 2005;18(4):719-756. alence of antibodies to spotted fever group rickettsiae in human
7. Goddard J. Historical and recent evidence for close relationships beings and dogs from an endemic area of Mediterranean spotted
among Rickettsia parkeri, R. conorii, R. africae, and R. sibirica: impli- fever in Catalonia, Spain. Eur J Epidemiol. 1998;14(4):395-398.
cations for rickettsial taxonomy. J Vector Ecol. 2009;34(2):238-242. 28. Beeler E, Abramowicz KF, Zambrano ML, et al. A focus of dogs
8. Garcia-Garcia JC, Portillo A, Nunez MJ, et  al. A patient from and Rickettsia massiliae–infected Rhipicephalus sanguineus in Cal-
Argentina infected with Rickettsia massiliae. Am J Trop Med Hyg. ifornia. Am J Trop Med Hyg. 2011;84(2):244-249.
2010;82(4):691-692. 29. Sexton DJ, Banks J, Graves S, et  al. Prevalence of antibodies to
9. Cragun WC, Bartlett BL, Ellis MW, et al. The expanding spectrum spotted fever group rickettsiae in dogs from southeastern Australia.
of eschar-associated rickettsioses in the United States. Arch Derma- Am J Trop Med Hyg. 1991;45(2):243-248.
tol. 2010;146(6):641-648. 30. Satoh H, Tsuneki A, Inokuma H, et al. Seroprevalence of antibod-
10. Nicholson WL, Paddock CD, Demma L, et  al. Rocky Mountain ies against spotted fever group Rickettsia among dogs and humans
spotted fever in Arizona: documentation of heavy environmental in Okinawa, Japan. Microbiol Immunol. 2001;45(1):85-87.
infestations of Rhipicephalus sanguineus at an endemic site. Ann N 31. Inokuma H, Matsuda H, Sakamoto L, et  al. Evaluation of Rick-
Y Acad Sci. 2006;1078:338-341. ettsia japonica pathogenesis and reservoir potential in dogs by
11. Demma LJ, Eremeeva M, Nicholson WL, et  al. An outbreak of experimental inoculation and epidemiologic survey. Clin Vaccine
Rocky Mountain spotted fever associated with a novel tick vector, Immunol. 2011;18(1):161-166.
Rhipicephalus sanguineus, in Arizona, 2004: preliminary report. 32. Greene CE, Breitschwerdt EB. Rocky Mountain spotted fever,
Ann N Y Acad Sci. 2006;1078:342-343. murine typhus-like disease, rickettsialpox, typhus, and Q fever. In:
12. Parola P, Socolovschi C, Jeanjean L, et al. Warmer weather linked Greene CE, ed. Infectious Diseases of the Dog and Cat. 6th ed.
to tick attack and emergence of severe rickettsioses. PLoS Negl Philadelphia, PA: Saunders Elsevier; 2006:232-241.
Trop Dis. 2008;2(11):e338. 33. Bayliss DB, Morris AK, Horta MC, et al. Prevalence of Rickettsia
13. Parola P, Labruna MB, Raoult D. Tick-borne rickettsioses in Amer- species antibodies and Rickettsia species DNA in the blood of cats
ica: unanswered questions and emerging diseases. Curr Infect Dis with and without fever. J Feline Med Surg. 2009;11(4):266-270.
Rep. 2009;11(1):40-50. 34. Warner RD, Marsh WW. Rocky Mountain spotted fever. J Am Vet
14. Solano-Gallego L, Kidd L, Trotta M, et al. Febrile illness associated Med Assoc. 2002;221(10):1413-1417.
with Rickettsia conorii infection in dogs from Sicily. Emerg Infect 35. Guedes E, Leite RC, Prata MC, et al. Detection of Rickettsia rick-
Dis. 2006;12(12):1985-1988. ettsii in the tick Amblyomma cajennense in a new Brazilian spotted
15. Keenan KP, Buhles Jr WC, Huxsoll DL, et al. Studies on the pathogen- fever-endemic area in the state of Minas Gerais. Mem Inst Oswaldo
esis of Rickettsia rickettsii in the dog: clinical and clinicopathologic Cruz. 2005;100(8):841-845.
changes of experimental infection. Am J Vet Res. 1977;38(6):851-856. 36. Breitschwerdt EB, Hegarty BC, Maggi RG, Lantos PM, Aslett DM,
16. Keenan KP, Buhles Jr WC, Huxsoll DL, et al. Pathogenesis of infec- Bradley JM. Rickettsia rickettsii Transmission by a Lone Star Tick,
tion with Rickettsia rickettsii in the dog: a disease model for Rocky North Carolina. Emerg Infect Dis. 2011;17(5):873-875.
Mountain spotted fever. J Infect Dis. 1977;135(6):911-917. 37. Demma LJ, Traeger MS, Nicholson WL, et  al. Rocky Mountain
17. Gasser AM, Birkenheuer AJ, Breitschwerdt EB. Canine Rocky spotted fever from an unexpected tick vector in Arizona. N Engl J
Mountain spotted fever: a retrospective study of 30 cases. J Am Med. 2005;353(6):587-594.
Anim Hosp Assoc. 2001;37(1):41-48. 38. Piranda EM, Faccini JL, Pinter A, et al. Experimental infection of
18. Greene CE, Burgdorfer W, Cavagnolo R, et  al. Rocky Mountain Rhipicephalus sanguineus ticks with the bacterium Rickettsia rick-
spotted fever in dogs and its differentiation from canine ehrlichio- ettsii, using experimentally infected dogs. Vector Borne Zoonotic
sis. J Am Vet Med Assoc. 1985;186(5):465-472. Dis. 2011;11(1):29-36.
310 SECTION 2  Bacterial Diseases

39. Chen LF, Sexton DJ. What’s new in Rocky Mountain spotted 62. Breitschwerdt EB, Papich MG, Hegarty BC, et al. Efficacy of dox-
fever? Infect Dis Clin North Am. 2008;22(3):415-432:vii-viii. ycycline, azithromycin, or trovafloxacin for treatment of experi-
40. Dantas-Torres F. Rocky Mountain spotted fever. Lancet Infect Dis. mental Rocky Mountain spotted fever in dogs. Antimicrob Agents
2007;7(11):724-732. Chemother. 1999;43(4):813-821.
41. Kidd LB. Transmission times and prevention of tick-borne diseases 63. Piranda EM, Faccini JL, Pinter A, et  al. Experimental infec-
in dogs. Compend Contin Educ Pract Vet. 2003;25(10):742-751. tion of dogs with a Brazilian strain of Rickettsia rickettsii:
42. Dantas-Torres F. Biology and ecology of the brown dog tick. Rhipi- clinical and laboratory findings. Mem Inst Oswaldo Cruz.
cephalus sanguineus. Parasit Vectors. 2010;3:26. 2008;103(7):696-701.
43. Norment BR, Burgdorfer W. Susceptibility and reservoir poten- 64. Kordick SK, Breitschwerdt EB, Hegarty BC, et al. Coinfection with
tial of the dog to spotted fever-group rickettsiae. Am J Vet Res. multiple tick-borne pathogens in a Walker Hound kennel in North
1984;45(9):1706-1710. Carolina. J Clin Microbiol. 1999;37(8):2631-2638.
44. Wikswo ME, Hu R, Metzger ME, Eremeeva ME. Detection of Rick- 65. Greene CE. Infectious Diseases of the Dog and Cat. 3rd ed. St.
ettsia rickettsii and Bartonella henselae in Rhipicephalus sanguin- Louis, MO: Saunders Elsevier; 2006.
eus ticks from California. J Med Entomol. 2007;44(1):158-162. 66. Ober CP, Spaulding K, Breitschwerdt EB, et  al. Orchitis in two
45. Stromdahl EY, Jiang J, Vince M, Richards AL. Infrequency of Rick- dogs with Rocky Mountain spotted fever. Vet Radiol Ultrasound.
ettsia rickettsii in Dermacentor variabilis removed from humans, 2004;45(5):458-465.
with comments on the role of other human-biting ticks associated 67. Grindem CB, Breitschwerdt EB, Perkins PC, et  al. Platelet-asso-
with spotted fever group Rickettsiae in the United States. Vector ciated immunoglobulin (antiplatelet antibody) in canine Rocky
Borne Zoonotic Dis. 2011;11(7):969-977. Mountain spotted fever and ehrlichiosis. J Am Anim Hosp Assoc.
46. Chapman AS, Bakken JS, Folk SM, et al. Diagnosis and manage- 1999;35(1):56-61.
ment of tickborne rickettsial diseases: Rocky Mountain spotted 68. Mikszewski JS, Vite CH. Central nervous system dysfunction asso-
fever, ehrlichioses, and anaplasmosis—United States: a practical ciated with Rocky Mountain spotted fever infection in five dogs.
guide for physicians and other health-care and public health pro- J Am Anim Hosp Assoc. 2005;41(4):259-266.
fessionals. MMWR Recomm Rep. 2006;55(RR-4):1-27. 69. Drost WT, Berry CR, Breitschwerdt EB, Davidson MG. Thoracic
47. Centers for Disease Control and Prevention. Rocky Mountain radiographic findings in dogs infected with Rickettsia rickettsii. Vet
Spotted Fever, Statistics and Epidemiology. http://www.cdc.gov Radiol Ultrasound. 1997;38(4):260-266.
/rmsf/stats:Last accessed May 27, 2012. 70. Breitschwerdt EB, Levy MG, Davidson MG, et al. Kinetics of IgM
48. Moncayo AC, Cohen SB, Fritzen CM, et  al. Absence of and IgG responses to experimental and naturally acquired Rickettsia
­Rickettsia rickettsii and occurrence of other spotted fever rickettsii infection in dogs. Am J Vet Res. 1990;51(8):1312-1316.
group rickettsiae in ticks from Tennessee. Am J Trop Med Hyg. 71. Nicholson WL, Allen KE, McQuiston JH, et al. The increasing recog-
2010;83(3):653-657. nition of rickettsial pathogens in dogs and people. Trends Parasitol.
49. Raoult D, Parola P. Rocky Mountain spotted fever in the USA: a 2010;26(4):205-212.
benign disease or a common diagnostic error? Lancet Infect Dis. 72. Breitschwerdt EB, Moncol DJ, Corbett WT, et  al. Antibodies to
2008;8(10):587-589. spotted fever-group rickettsiae in dogs in North Carolina. Am J Vet
50. Diniz PPVP, unpublished data. 2012. Res. 1987;48(10):1436-1440.
51. Eremeeva ME, Zambrano ML, Anaya L, et  al. Rickettsia rick- 73. Solano-Gallego L, Bradley J, Hegarty B, et al. Bartonella henselae
ettsii in Rhipicephalus ticks, Mexicali. Mexico. J Med Entomol. IgG antibodies are prevalent in dogs from southeastern USA. Vet
2011;48(2):418-421. Res. 2004;35(5):585-595.
52. Tinoco-Gracia L, Quiroz-Romero H, Quintero-Martinez MT, 74. Kidd L, Maggi R, Diniz PP, et  al. Evaluation of conventional
et  al. Prevalence of Rhipicephalus sanguineus ticks on dogs in a and real-time PCR assays for detection and differentiation of
region on the Mexico-USA border. Vet Rec. 2009;164(2):59-61. spotted fever group Rickettsia in dog blood. Vet Microbiol.
53. Fritz CL, Kriner P, Garcia D, et  al. Tick infestation and spotted 2008;129(3-4):294-303.
fever group Rickettsia in shelter dogs, California, 2009. Zoonoses 75. Davidson MG, Breitschwerdt EB, Walker DH, et  al. Identifica-
Public Health. 2012;59(1):4-7. tion of rickettsiae in cutaneous biopsy specimens from dogs with
54. Telford SR. Status of the “East Side hypothesis” (transovarial inter- experimental Rocky Mountain spotted fever. J Vet Intern Med.
ference) 25 years later. Ann N Y Acad Sci. 2009;1166:144-150. 1989;3(1):8-11.
55. Sahni SK, Rydkina E. Host-cell interactions with pathogenic Rick- 76. Breitschwerdt EB, Davidson MG, Aucoin DP, et  al. Efficacy of
ettsia species. Future Microbiol. 2009;4(3):323-339. chloramphenicol, enrofloxacin, and tetracycline for treatment of
56. Walker DH, Valbuena GA, Olano JP. Pathogenic mechanisms of experimental Rocky Mountain spotted fever in dogs. Antimicrob
diseases caused by Rickettsia. Ann N Y Acad Sci. 2003;990:1-11. Agents Chemother. 1991;35(11):2375-2381.
57. Walker DH. Rickettsiae and rickettsial infections: the current state 77. Neer TM, Eddlestone SM, Gaunt SD, Corstvet RE. Efficacy of
of knowledge. Clin Infect Dis. 2007;45(suppl 1):S39-S44. enrofloxacin for the treatment of experimentally induced Ehrlichia
58. Balraj P, Renesto P, Raoult D. Advances in rickettsia pathogenicity. canis infection. J Vet Intern Med. 1999;13(5):501-504.
Ann N Y Acad Sci. 2009;1166:94-105. 78. Breitschwerdt EB, Davidson MG, Hegarty BC, et al. Prednisolone
59. Eremeeva ME, Liang Z, Paddock C, et al. Rickettsia rickettsii infec- at anti-inflammatory or immunosuppressive dosages in conjunc-
tion in the pine vole, Microtus pinetorum: kinetics of infection and tion with doxycycline does not potentiate the severity of Rick-
quantitation of antioxidant enzyme gene expression by RT-PCR. ettsia rickettsii infection in dogs. Antimicrob Agents Chemother.
Ann N Y Acad Sci. 2003;990:468-473. 1997;41(1):141-147.
60. Cummiskey JM, Simon LM, Theodore J, Ryan US, Robin ED. 79. Low RM, Holm JL. Canine Rocky Mountain spotted fever. Com-
Bioenergetic alterations in cultivated pulmonary artery and aortic pend Contin Educ Pract Vet. 2005;27(7):530-538.
endothelial cells exposed to normoxia and hypoxia. Exp Lung Res. 80. Walker DH. The realities of biodefense vaccines against Rickettsia.
1981;2(3):155-163. Vaccine. 2009;27(suppl 4):D52-D55.
61. Asahina T, Kashiwagi A, Nishio Y, et  al. Impaired activation 81. Bonilla D. Tick borne disease surveillance: spotted fever group
of glucose oxidation and NADPH supply in human endothe- Rickettsia. Calif Dept Public Health Vector Borne Dis Bull.
lial cells exposed to H2O2 in high-glucose medium. Diabetes. 2009;3(2):4.
1995;44(5):520-526.
CHAPTER 31

Salmon Poisoning Disease


Jane E. Sykes

the distribution of an aquatic snail, Oxytrema silicula (also known


Overview of Salmon Poisoning Disease as Juga silicula), which is an intermediate host of the trematode
First Described: Northwestern Oregon, United States, 1814 (Figure 31-1). The snail is prevalent in coastal streams. A similar
(Astoria)1 disease has been described in dogs from Brazil.6,7 Two related
Cause: Neorickettsia helminthoeca, a gram-negative, obli- yet antigenically distinct organisms have been isolated from
gately intracellular bacteria that belongs to the family dogs with SPD, N. helminthoeca and the Elokomin fluke fever
Anaplasmataceae (EFF) agent.8-12 The EFF agent may be a less pathogenic strain of
N. helminthoeca.
Affected Hosts: Dogs, foxes, coyotes, raccoons, captive bears
Dogs usually become infected with N. helminthoeca when
Intermediate Hosts: Aquatic snails, fish (especially salmonids) they ingest encysted trematode metacercariae within uncooked
Geographic Distribution: Coastal areas of Washington, ­Oregon, or undercooked freshwater fish, most commonly salmonid fish.
northern California, British Columbia, Brazil (2004) The Pacific giant salamander is also a competent second inter-
mediate host. Ingestion of encysted metacercariae is followed
Route of Transmission: Ingestion of the infected trematode
by maturation of the trematode, which feeds on the intestinal
vector Nanophyetus salmincola, most often in salmonid
mucosa and inoculates the rickettsia into the host. The fluke
fish
has an oral sucker and a ventral sucker, which are used to grasp
Major Clinical Signs: Fever, lethargy, anorexia, vomiting, diar- host intestinal tissue, although the fluke itself does not cause
rhea, lymphadenomegaly extensive damage to the intestinal wall.13 Infected trematode
Differential Diagnoses: Lymphoma, canine parvovirus infec- ova are shed in the stool for 60 to 250 days.14 After several
tion, canine distemper virus infection, granulocytic months, miracidia develop within the eggs, hatch, swim away,
anaplasmosis, ehrlichiosis, leptospirosis, septic shock, and penetrate the snail. They then develop into rediae, each
disseminated fungal disease (especially cryptococco- of which give rise to many free-swimming cercariae. Thou-
sis), hemorrhagic gastroenteritis, dietary indiscretion, sands of cercariae are released intermittently from the snail.
gastrointestinal foreign body, pancreatitis, hypoadreno- These rapidly penetrate the skin of a fish (or are ingested by
corticism, inflammatory bowel disease. Appropriate anti- the fish), and encyst throughout the fish as metacercariae
microbial treatment with doxycycline must begin before
the diagnosis is confirmed by laboratory testing. Misdi-
agnosis and delayed or inappropriate antimicrobial drug
therapy increase morbidity and mortality.
Human Health Significance: Nanophyetus salmincola can
cause gastrointestinal disturbances and eosinophilia
in humans, but N. helminthoeca does not cause human
disease.

Etiologic Agent and Epidemiology


Salmon poisoning disease (SPD) is caused by the rickettsial
pathogen Neorickettsia helminthoeca, which, like other Neor-
ickettsia spp., resides within a trematode (fluke) vector for the
course of the trematode’s life cycle. The trematode vector of
N. helminthoeca is Nanophyetus salmincola, thought to be the
most common trematode species endemic to the United States.2
The disease occurs in dogs, foxes, and coyotes and has also been
reported in captive bears, but does not occur in domestic cats.3-5
SPD is geographically restricted to coastal regions of northern FIGURE 31-1  Geographic distribution of salmon poisoning disease in dogs from
California, Oregon, and Washington in the United States, and the United States. The distribution also extends into the southern portion of ­British
southern British Columbia in Canada (Figure 31-1). This reflects Columbia.

311
312 SECTION 2  Bacterial Diseases

Adult fluke within


intestinal tract
feeds on lining
and injects
bacteria
Eggs
in feces
Metacercaria
encysted in
salmonid fish
A
Miracidium

Snail

Cercaria swims to and


penetrates salmonid fish B
FIGURE 31-2  Life cycle of Nanophyetus salmincola, the trematode vector of salmon FIGURE 31-3  A, “Salt grain” appearance of metacercariae of Nanophyetus salmincola
poisoning disease. The fluke harbors the rickettsial organism throughout its lifecycle. (arrows) in the kidneys of a Chinook salmon. (Courtesy Craig Banner, Oregon Department
of Fish and Wildlife.) B, Wet mount squash of fresh kidney material from a Chinook salmon
showing a metacercaria of N. salmincola. Bar = 100 µm. (Courtesy Dr. Ronald Hedrick, Uni-
versity of California, Davis.)
(Figures 31-2 and 31-3). Infected fish may contain more than
1000 metacercariae. Infected fish may have visible damage to
their skin, and the infection may also interfere with their swim-
ming activity.15 Dog-to-dog transmission of N. helminthoeca
BOX 31-1
has been demonstrated experimentally by mechanical aerosol-
Signs in the Clinical History of 29 Dogs with Salmon
ization or rectal administration of lymph node suspensions or
homogenates of rectal mucosa.16
Poisoning Disease
Intact male dogs and Labrador retrievers appear to be over-
represented among dogs with SPD,17 possibly because they are Inappetence or anorexia: 100%
popular with individuals engaged in fishing activities, although Lethargy: 86%
dogs of any sex and breed can be affected. Dogs of any age Vomiting: 86%
are affected; the median age in one study was 3 years.17 Dogs Diarrhea: 72%
develop SPD at any time of year. Occasionally dogs from areas Hematochezia: 38%
not endemic for SPD develop disease after they ingest fish that is Weight loss: 28%
imported from endemic areas.18 Melena: 21%
Most but not all infected dogs have a history of access to Polyuria and polydipsia: 3%
or consumption of raw or improperly cooked fish.17,19 Con-
sumption of any part of the fish, including the entrails and
skin, may result in SPD. Supermarket-bought salmon may also
be infected.17 Infection has also been reported after swimming infected dogs.17 Terminally ill dogs may be hypothermic. Vir-
activity, without apparent exposure to fish.17 tually all dogs develop inappetence or anorexia, and lethargy
is also common. Vomiting occurs in more than 80% of dogs,
Clinical Features and rarely, hematemesis is present.17 Diarrhea is also common
(>70% of dogs) and may be semiformed to liquid in consistency,
Signs and Their Pathogenesis sometimes containing frank blood, melena, and, less commonly,
N. helminthoeca replicates within macrophages. This results in a mucus.17 Occasionally, a history of diarrhea and vomiting is
granulomatous inflammatory response within the stomach, intes- absent. Other signs include weight loss and increased thirst and
tines, lymph nodes, and spleen. Granulomatous meningitis has urination. Neurologic signs that include mental obtundation,
also been detected in dogs with SPD.20,21 The incubation period myoclonic twitching, seizures, and apparent neck pain have been
ranges from 2 to 14 days (median, 7 days), but may be as long as reported in fewer than 20% of affected dogs, a primary differen-
33 days.22 The severity of illness varies considerably (Box 31-1). tial diagnosis being canine distemper virus infection.17
Dogs with peracute infection may be found dead.17 A marked
febrile response is commonly detected (>70% of cases) (Box Physical Examination Findings
31-2) and may reach 107.6°F (42°C).17A median rectal tempera- Dehydration is frequently noted on physical examination
ture of 105°F (40.6°C) was reported in one series of naturally and may be severe. More than 70% of dogs have peripheral
CHAPTER 31  Salmon Poisoning Disease 313

lymphadenomegaly, which may be generalized or involve only occurs as a result of gastrointestinal hemorrhage. More com-
some peripheral lymph nodes (see Box 31-2). Lymph nodes may monly, the CBC shows evidence of lymphopenia, thrombo-
be up to 4 cm in diameter and firm on palpation. Lymphad- cytopenia, or neutrophilia, frequently with bandemia (Table
enomegaly may be absent occasionally, sometimes as a result 31-1). Uncommonly, neutropenia occurs.17 Toxic changes
of lymph node necrosis. Abdominal palpation may reveal pain, may be present within neutrophils, and immature monocytes
and splenomegaly may also be detected. Other abnormalities may occasionally be found in the peripheral blood. Despite
include tachypnea or labored respirations, tachycardia, scleral the association with helminthiasis, eosinophilia has not been
or conjunctival injection, and edema that involves the limbs, documented.
face, or cervical region.17 Mucosal pallor, tachycardia, and
weak pulses may be present in dogs with hypovolemic shock. Serum Chemistry Profile
Cardiac arrhythmias such as ventricular premature contractions Hyponatremia and hypokalemia are the most common elec-
have also been noted, and there may be signs of hemorrhage trolyte abnormalities in dogs with SPD and result from gas-
such as epistaxis and hyphema. Rectal examination may indi- trointestinal losses that occur with vomiting and diarrhea
cate the presence of liquid stool, sometimes accompanied by (Table 31-2). Low ionized calcium concentrations may be
fresh blood or melena. detected in some dogs, possibly as a result of decreased intes-
tinal absorption. Hypoalbuminemia is present in most (>80%)
Diagnosis dogs. Hypoglobulinemia and hypocholesterolemia occur in
fewer than 25% of dogs with SPD, but may become pro-
Laboratory Abnormalities found. Abnormal liver enzyme activities may be present, the
Complete Blood Count most common of which is increased ALP activity,17,20 but
Thrombocytopenia is present in 90% of affected dogs. Ane- increases in the activity of ALT and AST may also occur.
mia is present in up to 40% of dogs with SPD and generally Hyperbilirubinemia is present in fewer than 20% of dogs. Azo-
temia can be evident in dogs with severe dehydration.

BOX 31-2 Urinalysis


Bilirubinuria and proteinuria are frequently detected on urinaly-
Physical Examination Findings in 29 Dogs with Salmon sis in dogs with SPD.17 Uncommonly, cylindruria and glucos-
Poisoning Disease uria are present, perhaps as a result of renal injury secondary
to impaired renal perfusion. Microscopic hematuria occurs in
some dogs, possibly reflecting underlying coagulopathies that
Peripheral lymphadenomegaly: 74%
can occur in dogs with SPD.
Fever: 73%
Dehydration: 65%
Abdominal pain: 28%
Clotting Function
Evaluation of clotting function in some dogs with SPD shows
Tachypnea or labored respirations: 24%
evidence of disseminated intravascular coagulation. Abnormali-
Tachycardia: 24%
ties include increased PT or APTT, increased fibrin degradation
Neurologic signs: 17%
products, and decreased antithrombin activity.17
Weak pulses: 17%
Splenomegaly: 14% Diagnostic Imaging
Scleral injection: 14%
Mucosal pallor: 10%
Plain Radiography
Abdominal radiographs may be unremarkable or reveal fluid-
Hypothermia: 5%
filled small intestines; decreased serosal detail; and less fre-
quently, hepatomegaly or splenomegaly. Thoracic radiographs

TABLE 31-1
Hematologic Findings in 29 Dogs with Salmon Poisoning Disease
Percent below Percent within Percent above Range for Dogs Number
Reference the Reference the Reference the Reference with Salmon of Dogs
Test Range Range Range Range Poisoning Disease Tested
Hematocrit (%) 40-55 39 61 0 22-53 23
Neutrophils (cells/µL) 3000-10,500 4 53 43 728-26,224 23
Band neutrophils (cells/µL) Rare 0 40 60 0-6825 20
Monocytes (cells/µL) 150-1200 9 61 30 42-3290 23
Lymphocytes (cells/µL) 1000-4000 78 22 0 0-2070 23
Eosinophils (cells/µL) 0-1500 0 100 0 0-690 21
Platelets (cells/µL) 150,000-400,000 90 0 10 10,000-446,000 20
314 SECTION 2  Bacterial Diseases

TABLE 31-2
Findings on Serum Biochemistry Analysis in 29 Dogs with Salmon Poisoning Disease
Percent below Percent within Percent above Range for Dogs
Reference the Reference the Reference the Reference with Salmon Number of
Test Range Range Range Range Poisoning Disease Dogs Tested
Sodium (mmol/L) 145-154 59 36 5 133-161 22
Potassium (mmol/L) 4.1-5.3 41 59 0 3.7-5.0 22
Chloride (mmol/L) 105-116 9 77 14 98-118 22
Bicarbonate (mmol/L) 16-26 20 15 65 11-28 20
Calcium (mg/dL) 9.9-11.4 83 17 0 7.2-10.9 23
Phosphorus (mg/dL) 3.0-6.2 9 74 17 2.7-13.0 23
Creatinine (mg/dL) 0.5-1.6 4 4 13 0.4-3.3 23
BUN (mg/dL) 8-31 0 83 17 8-110 23
Albumin (g/dL) 2.9-4.2 83 17 0 0.9-3.8 23
Globulin (g/dL) 2.3-4.4 26 70 4 1.6-4.7 23
Cholesterol (mg/dL) 135-345 22 69 9 79-408 23
Total bilirubin (mg/dL) 0-0.4 0 83 17 0-3.1 23
ALT (U/L) 19-70 4 74 22 17-1,443 23
ALP (U/L) 15-127 0 39 61 29-985 23

loops, sometimes with wall thickening, corrugation, hyper-


motility, or hypomotility, may be documented. Uncommonly,
hepatomegaly or a small amount of free peritoneal fluid is
identified.

Microbiologic Tests
Diagnostic assays for SPD are summarized in Table 31-3.

Diagnosis Using Fecal Examination


SPD can be diagnosed when operculated trematode eggs are
found in fecal specimens of dogs with consistent clinical signs,
which appear within 5 to 8 days after the ingestion of infected
fish. Although the sensitivity of centrifugal zinc sulfate fecal
flotation for detection of N. salmincola ova in dogs with SPD
is similar to that of fecal sedimentation, some dogs that test
FIGURE 31-4  Enlarged and hypoechoic abdominal (medial iliac) lymph nodes as negative by one method test positive using the other, so the sen-
detected using abdominal sonographic examination in a 9-year-old male German shep- sitivity of fecal examination for diagnosis is maximized by per-
herd dog with salmon poisoning disease. forming both centrifugal zinc sulfate fecal flotations and fecal
sedimentation together. In one study, 93% of dogs with SPD
tested positive for N. salmincola ova using a combination of
are typically unremarkable or show reduction in size of the fecal flotation and fecal sedimentation.17 False negatives may
intrathoracic vasculature, consistent with hypovolemia. occur when the duration of illness is shorter than the prepatent
period of N. salmincola or when light fluke burdens are pres-
Sonographic Findings ent. The finding of fluke ova on fecal examination may not be
Abdominal ultrasound examination of dogs with SPD often diagnostic of SPD, because dogs may be infected with trema-
shows mild or moderate generalized abdominal or mesenteric todes that do not harbor the rickettsia, and ova may be shed
lymphadenomegaly, as well as splenomegaly with a mottled for months after recovery. Nevertheless, N. salmincola ova were
splenic echotexture. Some dogs that lack peripheral lymphad- detected in only 0.2% of more than 1800 fecal flotations in dogs
enomegaly have abdominal lymphadenomegaly on ultrasound seen at a teaching hospital in an endemic area, and all positive
examination,17 and ultrasound-guided aspiration of enlarged test results were in dogs suspected to have SPD. Thus, the speci-
abdominal lymph nodes can be helpful for diagnosis in dogs ficity of fecal examination for a diagnosis of SPD appears to be
that lack peripheral lymphadenomegaly. Lymph nodes develop extremely high.17
a rounded appearance with a hypoechoic or hypoechoic Centrifugal zinc sulfate fecal flotation may reveal co-infec-
mottled echogenicity (Figure 31-4). Fluid-distended intestinal tions with other gastrointestinal parasites, which can complicate
CHAPTER 31  Salmon Poisoning Disease 315

the clinical picture. Co-infections with Dipylidium caninum and dogs.17 Real-time PCR assays for N. helminthoeca are avail-
Trichuris vulpis have been reported in dogs with SPD.17 able from some veterinary molecular diagnostic laboratories
and can be performed on blood, lymph-node, or splenic aspi-
Cytologic Diagnosis rates and possibly fecal specimens from dogs with SPD in
In addition to fecal examination for trematode ova, SPD can be order to confirm the diagnosis. More research is required to
diagnosed following cytologic examination of peripheral lymph understand the sensitivity and specificity of PCR assays for
node aspirates, or ultrasound-guided aspirates of abdominal diagnosing SPD in dogs, including the optimum specimen type
lymph nodes or the spleen. Cytologic findings consist of histio- for testing.
cytic hyperplasia and lymphoid reactivity. Rickettsial organ-
isms are 0.3−µm cocci or coccobacilli and may be seen in large
numbers within the cytoplasm of macrophages. They resemble
granular to amorphous material of a uniform blue color using
Wright’s stain, occasionally forming loose clusters of organ-
isms (morulae) (Figure 31-5).17,22,23 In as many as one quarter
to one third of dogs, rickettsial organisms are not identified.
The presence of histiocytic hyperplasia should alert the clini-
cian to the possibility of SPD. Organisms may also be absent in
dogs with a recent history of antimicrobial therapy.

Serologic Diagnosis
Serologic tests for N. helminthoeca antibodies or antigen are
not available on a commercial basis for veterinary practitio-
ners. Antibodies to N. helminthoeca can cross-react sero-
logically with those of Ehrlichia spp., so it is possible that
dogs with SPD might test positive using serologic tests for
Ehrlichia spp. (provided enough time had elapsed for anti-
body formation to occur).24 In one study, most dogs with
SPD that were tested for antibodies against other rickettsial
pathogens did not seroreact to Ehrlichia canis or Anaplasma
phagocytophilum.17

Molecular Diagnosis Using the Polymerase Chain Reaction FIGURE 31-5  Lymph node aspirate from a dog with salmon poisoning disease. Note
Specific PCR assays have been used to detect the DNA of the large histiocyte that contains granular coccoid organisms consistent with Neorickettsia
Neorickettsia helminthoeca in clinical specimens from affected helminthoeca.

TABLE 31-3
Diagnostic Assays Available for Salmon Poisoning Disease in Dogs
Assay Specimen Type Target Performance
Fecal sedimentation Feces Nanophyetus salmincola Specificity nears 100% when performed by a parasi-
combined with zinc ova tologist. Sensitivity probably >90%. Eggs are light
sulfate centrifugal brown, ovoid, operculate at one end and measure
fecal flotation 0.087 to 0.097 mm × 0.038 to 0.055 mm.
Cytology Lymph-node or Detection of N
­ eorickettsia Sensitivity >70% when performed by a veterinary
splenic aspirates helminthoeca within clinical pathologist. Sensitivity may be improved by
­macrophages examination of aspirates from multiple lymph node.
Organisms within macrophages may be confused
with debris or hemosiderin. Other changes include
lymphocytic reactivity and histiocytic hyperplasia.
Histopathology Biopsy or ­necropsy N. helminthoeca within Organisms may be stained with Giemsa. Antimicro-
specimens, espe- ­macrophages bial therapy can lead to false-negative results.
cially lymph nodes,
spleen, gastrointes-
tinal tissues
PCR Blood, lymph- N. helminthoeca DNA Well validated assays that are specific for
node or splenic N. ­helminthoeca are not available commercially.
aspirates, tissue Optimum specimen type unknown. Antimicrobial
specimens, feces therapy may lead to negative PCR results.
316 SECTION 2  Bacterial Diseases

Pathologic Findings
Gross Pathologic Findings
Gross necropsy findings in dogs with SPD consist of enlarge-
ment of lymphoid tissues, including the tonsils, thymus,
lymph nodes, and spleen. Lymph nodes may be yellowish and
edematous and may contain white foci that represent foci of
granulomatous inflammation. Petechial hemorrhages may be
noted on the lymph nodes, the gastrointestinal tract, pan-
creas, gallbladder, and urinary bladder. The gastrointestinal
tract may be thickened and contain white foci. The lumen of
the intestinal tract may contain free blood (Figure 31-6, A).
Flukes are only 0.8 to 1.1 mm long and 0.3 to 0.5 mm wide25
and may not always be visible grossly.

Histopathologic Findings A
Lymphoid tissues show marked depletion of lymphoid fol-
licles, and subcapsular and medullary sinuses are filled with
histiocytes, the cytoplasm of which can contain numerous
rickettsial organisms, which occasionally form clusters within
vacuoles. Organisms are readily appreciated using Giemsa
stain (see Figure 31-6, C). Lymph node and splenic necrosis
may also be present.17,22 Inflammatory nodules that contain
lymphocytes, plasma cells, and infected histiocytes expand the
lamina propria of the gastrointestinal tract and extend into
the submucosa. Parasites consistent with N. salmincola may
be found embedded in the mucosa, primarily within the duo-
denum (see Figure 31-6, B).16,21 Nevertheless, flukes are not
always found using histopathology in dogs that die from SPD,
even in the absence of anthelmintic therapy.
One dog that died of SPD had widespread and multiple
thromboses involving numerous organs with associated tis- B
sue infarction. Granulomatous meningitis has been described
at necropsy in dogs that were experimentally infected with
N. helminthoeca.20

Treatment and Prognosis


The treatment of choice for SPD is doxycycline, tetracycline,
or oxytetracycline, which should be given for a minimum of
7 days. Administration of tetracyclines is generally associated
with clinical improvement within 24 hours, and signs resolve
within 1 to 4 days.17 Dogs that are vomiting may require
treatment with parenteral tetracyclines. Clinical improvement
has also been reported following treatment with enrofloxacin
and parenteral trimethoprim-sulfamethoxazole. Antimicrobi-
als that do not appear to be effective include first-generation
cephalosporins, penicillins, aminoglycosides, chlorampheni-
col, and metronidazole.17 Although complete clinical recovery
C
may occur without anthelmintic treatment, the fluke infection FIGURE 31-6  A, Intestinal mucosa of an 8-month-old female spayed Maltese mix dog
should be treated with praziquantel after initial recovery from that died of salmon poisoning disease. The intestinal wall was thickened and the contents were
SPD (Table 31-4). dark red and mucoid. (Courtesy University of California, Davis Veterinary Anatomic Pathology
Dogs with mild signs or infrequent vomiting may respond Service.) B, Duodenum. A transverse section of an adult Nanophyetus fluke is embedded
well to treatment with oral tetracyclines alone, whereas those within the intestinal villi. H&E stain. Bar = 200 µm. C, Lymph node, medullary sinus from a dog
with salmon poisoning disease. Many histiocytes have intracytoplasmic clusters of coccobacilli
with more severe disease often require hospitalization for IV
less than 1 µm in diameter. Giemsa. Bar = 5 µm. (B and C from Sykes JE, Marks SL, Mapes S,
antimicrobial and crystalloid fluid therapy (see Table 31-4). et al. Salmon poisoning disease in dogs: 29 cases. J Vet Intern Med 2010;24[3]:504-513, 2010.)
Oral antimicrobial therapy can be continued once vomiting
ceases. The presence of severe hypovolemia or hypoalbumin-
emia may necessitate treatment with colloids such as fresh such as metoclopramide, maropitant, or ondansetron. Close
frozen plasma, dextrans, or hetastarch. Packed RBC or whole monitoring of hematocrit, albumin, electrolytes, acid-base
blood may be required for dogs with severe anemia. Treatment status, renal parameters, and coagulation parameters may be
with total or partial parenteral nutrition may also be required required in some severely affected dogs. Blood cultures should
if vomiting is persistent and does not respond to antiemetics be considered in dogs with severe hemorrhagic diarrhea and
CHAPTER 31  Salmon Poisoning Disease 317

Immunity and Vaccination


TABLE 31-4
Antimicrobials Used to Treat Salmon Poisoning Disease in Dogs Animals that recover from SPD are immune to reinfection with
the same strain of N. helminthoeca,22,26,27 but challenge with
Dose Interval Duration an alternate strain (then the EFF agent) results in disease, and
Drug (mg/kg) Route (hours) (days) antibodies fail to cross react with the alternate strain. This may
explain why SPD has been reported to occur more than once in
Doxycycline 5 PO, IV 12 7-14 some dogs.17,22 No vaccine is available.
Tetracycline 22* PO 8 7
7 IV 8 7 Prevention
Oxytetracycline 7-10* PO, IV 8 7
N. helminthoeca metacercariae are effectively destroyed by proper
Praziquantel† 10-30 PO 24 1-2
cooking or freezing of infected fish. Ingestion of raw fish by dogs in
*Reduce dose in renal failure; may cause teeth discoloration in young endemic areas should be discouraged, and dog owners in endemic
animals. Monitor for nephrotoxicity (cylindruria). areas should be educated about the disease. Dogs that have eaten
†For treatment of fluke infection. raw fish from an endemic area, or those swimming in rivers or
lakes in these areas, should be watched carefully for signs of leth-
argy, vomiting, or decreased appetite that occur within 2 weeks of
exposure. If multiple dogs are simultaneously exposed to a source
abnormal perfusion parameters, after which treatment with of N. helminthoeca, and one dog develops SPD, treatment of the
broad-spectrum parenteral antimicrobial drugs should be other dogs in the group with 7 days of doxycycline should be con-
commenced. sidered. Medical equipment used in the treatment of dogs with SPD
Prolonged hospitalization may be required for dogs that should not be reused on other patients without proper sterilization.
develop complications such as cardiac arrhythmias, intus-
susceptions, adverse reactions to tetracyclines, disseminated Public Health Aspects
intravascular coagulation, or septicemia.17,19 In a study of dogs
evaluated at a tertiary referral hospital, death or euthanasia Human infection with N. helminthoeca has not been reported.
occurred in 14% of 29 dogs with SPD.17 The mortality may be However, humans may become infected with the fluke, N. sal-
lower in dogs seen in primary care practice, providing appropri- mincola, after consumption of poorly cooked fish. Most humans
ate treatment can be administered. In general, early treatment are subclinically infected, although abdominal discomfort, diar-
is associated with an excellent prognosis. Without treatment, rhea, vomiting, weight loss, nausea, and peripheral eosinophilia
death can occur within 5 to 10 days. may occur.28

CASE EXAMPLE He was cryptorchid, and the abdominal testicle had been
removed when he was a young dog. The descended testicle
was still present.
Signalment: “Herman,” 9-year-old male German shepherd Physical Examination:
dog from Sacramento, CA (Figure 31-7). Body Weight: 51.5 kg
History: Herman’s owner reported a 2-day history of General: Quiet, alert and responsive. Ambulatory on all four
inappetence, lethargy, and one episode of vomiting. The dog limbs. T = 104.3°F (40.2°C), HR = 108 beats/min, RR = 60
had been drinking normally. There had been no diarrhea, breaths/min, mucous membranes pink, CRT = 1 s.
coughing, sneezing, or increased thirst and urination. The Integument: A full, shiny haircoat was present and there was
owner also reported that Herman ate turkey and chicken no evidence of ectoparasites.
that contained bones 3 days before the onset of illness. He Eyes, Ears, Nose, and Throat: No significant abnormalities
was normally fed a strict diet of rabbit- and potato-based were noted. A mild amount of brown waxy debris was
prescription dog food and grilled salmon because of atopic present within both ear canals.
dermatitis. Herman was sometimes fed salmon scraps from Musculoskeletal: A body condition score of 6/9 with
the local supermarket, which the owner grilled briefly. He symmetrical muscling was noted. Mild to moderate pelvic
had not traveled out of his local area. He was up to date limb weakness was identified.
on vaccinations, which included vaccines for distemper, Cardiovascular: Strong femoral pulses were noted. No murmurs
hepatitis, parvovirus, and rabies. or arrhythmias were detected on thoracic auscultation.
Current Medications: Lincomycin 29 mg/kg PO q12h; Respiratory: An increased respiratory rate with increased
trimeprazine tartrate 0.2 mg/kg PO q48h; prednisolone 0.1 abdominal effort was present.
mg/kg q48h for atopic dermatitis. Gastrointestinal and Genitourinary: Abdominal palpation
Other Medical History: Herman had been diagnosed with revealed a tense abdomen, with abdominal splinting, and
hip osteoarthritis using pelvic radiography 8 months earlier. splenomegaly. There was one descended testicle, which was
Controlled weight loss was the recommended treatment. smooth with no masses. No abnormalities were detected on

Continued
318 SECTION 2  Bacterial Diseases

GGT < 3 U/L (0-6 U/L)


Cholesterol 296 mg/dL (135-361 mg/dL)
Total bilirubin 0.4 mg/dL (0-0.2 mg/dL)
Magnesium 1.7 mg/dL (1.5-2.6 mg/dL).
Urinalysis: SGr 1.048; pH 6.0, 2+ protein (SSA), 1+ bilirubin, 1+
hemoprotein, no glucose, 0-1 WBC/HPF, 20-30 RBC/HPF, rare
ammonium biurate crystals, many lipid droplets
Imaging Findings:
Thoracic Radiographs: The cardiovascular and pulmonary
structures appeared within normal limits. A mild diffuse
bronchial pattern throughout all lung fields was considered
consistent with the age of the patient. Marked spondylosis
deformans was present throughout the thoracic spine.
Abdominal Ultrasound: The liver had a diffuse, coarse
echotexture. The spleen was markedly, diffusely enlarged
and had a diffuse hypoechoic, mottled echotexture. The
prostate gland was enlarged and had multiple small
anechoic cysts within its parenchyma. One testicle was
present in the scrotum and had a striated, heterogenous
echotexture. A second inguinal or intra-abdominal testis
was not observed. Multiple enlarged, hypoechoic inguinal
lymph nodes were present (see Figure 31-3). A large (2.5 cm),
round, hypoechoic mass with a focal area of mineralization
was present in the cranial abdomen caudal to the liver and
FIGURE 31-7  “Herman,” a 9-year-old male German shepherd dog with salmon most likely represented a lymph node. Multiple enlarged,
­poisoning disease. hypoechoic sublumbar lymph nodes were observed. The
splenic lymph nodes were enlarged and mildly hypoechoic.
rectal examination. Soft feces were present in the rectum, Cytology Findings: Cytology of ultrasound-guided splenic
and the prostate was palpable and symmetrical. aspirate: splenic stromal clumps contained increased
Lymph Nodes: Marked peripheral lymphadenopathy was numbers of plump histiocytes and many large clumps of
present. Mandibular, superficial cervical, and popliteal hemosiderin. Histiocytes were also present in moderate
lymph nodes were all 3-4 cm in diameter and firm. numbers throughout the smear, often in clumps, and often
Laboratory Findings: mildly vacuolated. They were occasionally erythrophagocytic
CBC: and often contain hemosiderin. Interpretation: moderate to
HCT 40.4% (40-55%) marked histiocytic inflammation.
MCV 65 fL (65-75 fL) Cytology of popliteal lymph node aspirate (see Figure 31-5):
MCHC 36.6 g/dL (33-36 g/dL) moderate reactive lymphoid hyperplasia with pyogranu-
WBC 4240 cells/µL (6000-13,000 cells/µL) lomatous inflammation was identified. Rare histiocytes
Neutrophils 3689 cells/µL (3000-10,500 cells/µL) were present that contained intracellular small coccoid
Lymphocytes 466 cells/µL (1000-4000 cells/µL) purple-blue structures that were organized individually
Monocytes 42 cells/µL (150-1200 cells/µL) and in clusters. These were consistent with Neorickettsia
Platelets 74,000 platelets/µL (150,000-400,000 platelets/µL). helminthoeca organisms.
Serum Chemistry Profile: Fecal examination: Fecal flotation: Negative for parasites.
Sodium 141 mmol/L (145-154 mmol/L) Fecal sedimentation: Nanophyetus salmincola ova, <1 per 10× field
Potassium 4.0 mmol/L (3.6-5.3 mmol/L) Diagnosis: Salmon poisoning disease
Chloride 112 mmol/L (108-118 mmol/L) Treatment: Doxycycline, 5 mg/kg PO q12h for 14 days. This
Bicarbonate 13 mmol/L (16-26 mmol/L) was associated with resolution of fever and inappetence
Phosphorus 2.8 mg/dL (3.0-6.2 mg/dL) within 24 hours of starting treatment. By 7 days after
Calcium 9.9 mg/dL (9.7-11.5 mg/dl) diagnosis, Herman’s owner reported that Herman was
BUN 15 mg/dL ( 5-21 mg/dL) completely back to his normal self.
Creatinine 1.1 mg/dL (0.3-1.2 mg/dL) Comments: This dog was diagnosed with SPD based on the
Glucose 86 mg/dL (64-123 mg/dL) presence of characteristic cytologic findings on examination
Total protein 6.9 g/dL (5.4-7.6 g/dL) of a lymph node aspirate, together with the detection of N.
Albumin 3.6 g/dL (3.0-4.4 g/dL) salmincola ova in the stool. The case was unusual because
Globulin 3.3 g/dL (1.8-3.9 g/dL) the dog was infected after ingestion of incompletely cooked
ALT 108 U/L (19-67 U/L) supermarket-bought salmon, which the dog had been fed
AST 109 U/L (19-42 U/L) for years without developing illness. Although the splenic
ALP 105 U/L (21-170 U/L) aspirate contained inflammatory cells typical of those seen
Creatine kinase 381 U/L (51-399 U/L) in SPD, organisms could not be convincingly documented.
CHAPTER 31  Salmon Poisoning Disease 319

SUGGESTED READINGS 13. Bennington E, Pratt I. The life history of the salmon-poisoning fluke,
Nanophyetus salmincola (Chapin). J Parasitol. 1960;46(1):91-100.
Mack RE, Bercovitch MG, Ling GV, et  al. Salmon poisoning disease 14. Foreyt WJ, Gorham JR, Green JS, et al. Salmon poisoning disease
complex in dogs—a review of 45 cases. Calif Vet. 1990;44:42-45. in juvenile coyotes: clinical evaluation and infectivity of metacer-
Sykes JE, Marks SL, Mapes S, et al. Salmon poisoning disease in dogs: cariae and rickettsiae. J Wild Dis. 1987;23(3):412-417.
29 cases. J Vet Intern Med. 2010;24(3):504-513. 15. Baldwin NL, Milleman RE, Knapp SE. “Salmon poisoning” dis-
ease. 3. Effect of experimental Nanophyetus salmincola infection
REFERENCES of the fish host. J Parasitol. 1967;53(3):556-564.
1. Millemann RE, Knapp SE. Biology of Nanophyetus salmincola and 16. Bosman DD, Farrell RK, Gorham JR. Non-endoparasite transmis-
“salmon poisoning disease.” In: Dawes B, ed. Advances in Parasi- sion of salmon poisoning disease of dogs. J Am Vet Med Assoc.
tology. vol. 8. New York, NY: Academic Press; 1970:1-39. 1970;156:1907-1910.
2. Harrell LW, Deardoff TL. Human nanophyetiasis: transmission by 17. Sykes JE, Marks SL, Mapes S, et al. Salmon poisoning disease in
handling naturally infected coho salmon (Oncorhynchus kisutch). dogs: 29 cases. J Vet Intern Med. 2010;24(3):504-513.
J Infect Dis. 1990;161(1):146-148. 18. Farrell RK, Dee JF, Ott RL. Salmon poisoning in a dog fed kippered
3. Foreyt WJ, Thorson S, Gorham JR. Experimental salmon poi- salmon. J Am Vet Med Assoc. 1968;152:370-371.
soning disease in juvenile coyotes (Canis latrans). J Wildl Dis. 19. Mack RE, Bercovitch MG, Ling GV, et al. Salmon poisoning disease
1982;18(2):159-162. complex in dogs—a review of 45 cases. Calif Vet. 1990;44:42-45.
4. Hedrick RP, Arnandi A, Manzer D. Nanophyetus salmincola: the 20. Philip CB, Hadlow WJ, Hughes LE. Neorickettsia helminthoeca,
trematode vector of Neorickettsia helmintheca. California Veteri- a new rickettsialike disease agent of dogs in western United
narian. 1990;44:18-22. States transmitted by a helminth. Proc 6th Int Congr Microbiol.
5. Gai JJ, Marks SL. Salmon poisoning disease in two Malayan sun 1953;4:70-82:Rome.
bears. J Am Vet Med Assoc. 2008;232(4):586-588. 21. Hadlow WJ. Neuropathology of experimental salmon poisoning of
6. Headley SA, Scorpio DG, Barat NC, et  al. Neorickettsia hel- dogs. Am J Vet Res. 1957;18:898-908.
minthoeca in dog. Brazil. Emerg Infect Dis. 2006;12:1303-1304. 22. Gorham JR, Foreyt WJ. Salmon poisoning disease. In: Greene CE,
7. Headley SA, Kano FS, Scorpio DG, et  al. Neorickettsia hel- ed. Clinical Microbiology and Infectious Diseases of the Dog and
minthoeca in Brazilian dogs: a cytopathological, histopatho- Cat. 1st ed. Philadelphia, PA: WB Saunders; 1984:538-544.
logical and immunohistochemical study. Clin Microbiol Infect. 23. Johns JL, Strasser JL, Zinkl JG, et al. Lymph node aspirate from a
2009;15(suppl 2):21-23. California wine-country dog. Vet Clin Pathol. 2006;35:243-246.
8. Cordy DR, Gorham JR. The pathology and etiology of salmon dis- 24. Rikihisa Y. Cross-reacting antigens between Neorickettsia hel-
ease in the dog and fox. Am J Pathol. 1950;26:617-637. minthoeca and Ehrlichia species, shown by immunofluorescence and
9. Farrell RK. Transmission of two rickettsia-like disease agents of Western immunoblotting. J Clin Microbiol. 1991;29:2024-2029.
dogs by endoparasites in northwestern USA. Proc 1st Int Congr 25. Witenburg G. On the anatomy and systematic position of the caus-
Parasitol. 1966:438. ative agent of so-called salmon poisoning. J Parasitol. 1932;18:258-263.
10. Farrell RK, Leader RW, Johnston SD. Differentiation of salmon 26. Donham CR, Simms BT, Miller FW. So-called salmon poisoning in
poisoning disease and Elokomin fluke fever: studies with the black dogs (progress report). J Am Vet Med Assoc. 1926;68:701-715.
bear (Ursus americanus). Am J Vet Res. 1973;34:919-922. 27. Sakawa H, Farrell RK, Mori M. Differentiation of salmon poison-
11. Kitao T, Farrell RK, Fukuda T. Differentiation of salmon poisoning ing disease and Elokomin fluke fever: complement fixation. Am
disease and Elokomin fluke fever: fluorescent antibody studies with J Vet Res. 1973;34:923-925.
Rickettsia sennetsu? Am J Vet Res. 1973;34:927-928. 28. Eastburn RL, Fritsche TR, Terhune Jr CA. Human intestinal
12. Frank DW, McGuire TC, Gorham JR, et  al. Cultivation of infection with Nanophyetus salmincola from salmonid fishes. Am
two species of Neorickettsia in canine monocytes. J Infect Dis. J Trop Med Hyg. 1987;36(3):586-591.
1974;129:257-262.
CHAPTER 32

Coxiellosis and Q Fever


Jane E. Sykes and Jacqueline M. Norris

milk is a less common and more controversial mode of trans-


Overview of Coxiellosis and Q Fever mission.6,7 The term Q fever stands for query fever, the name it
First Described: mid 1930s, Queensland, Australia, in slaugh- was given when it first emerged in Queensland, Australia, and
terhouse workers (Derrick)1 the underlying cause had not yet been identified. In humans,
Cause: Coxiella burnetii, an obligately intracellular gram-neg- acute Q fever is characterized by a febrile “flulike” illness, with
ative bacteria that belongs to the Family Coxiellaceae headaches, and sometimes pulmonary and hepatic involvement.
However, infection is initially asymptomatic in 60% of those
Affected Hosts: Humans are most susceptible; reproductive
infected.8 Chronic Q fever can follow acute Q fever or asymp-
disease can occur in farm animal species such as sheep,
tomatic infection and can be characterized by cardiac disease
goats, cattle, and horses, as well as cats and dogs
(endocarditis, pericarditis, myocarditis), fatigue syndromes,
Geographic Distribution: Worldwide, with the exception of skin rash, and neurologic, pulmonary, and hepatic disease. In
New Zealand avian species, systemic illness has been associated with infection
Major Clinical Signs: Dogs and cats are usually subclinically by other Coxiella-like organisms.9-11
infected. Abortion, premature birth, and stillbirth may Unlike other obligate intracellular pathogens, C. burnetii sur-
occur. vives prolonged periods in the environment, where it resists high
temperature, a variety of disinfectants, and ultraviolet light. The
Differential Diagnoses: Brucellosis, bartonellosis, herpesviral
organism exists in two forms: a small cell variant, which sur-
infections, leptospirosis, neosporosis, canine distemper,
vives in the environment, and a large cell variant, which repli-
and canine parvovirus infection. Noninfectious causes of
cates within cells. Survival of the organism for up to 10 months
pregnancy loss such as endocrinopathies, genetic disor-
on wool at 15°C to 20°C (59°F to 68°F) has been reported, and
ders, and nutritional deficiency should also be considered.
it can also survive in tissues that have been stored in formalin
Human Health Significance: Q fever in people results primar- for several months. It is inactivated by 5% hydrogen peroxide.12
ily from contact with infected sheep, goats, and cattle, or Although originally classified within the order Rickettsiales, the
indirect contact with surfaces contaminated with secre- availability of sequence information led to its reclassification as
tions or excretions of these animals. However, contact a gammaproteobacterium in the order Legionellales, which also
with infected companion animals can also lead to trans- includes Legionella species. C. burnetii undergoes phase varia-
mission. Q fever in people is most commonly an acute tion, which is associated with alterations in its cell wall com-
febrile illness, but pneumonia and chronic complica- ponents. The variants have nominally been called phase 1 and
tions such as endocarditis, granulomatous hepatitis, and phase 2.13 The changes in cell surface antigens and the creation
chronic fatigue can occur. of phase variants has clinical implications for serologic testing
due to the production of phase 1 and phase 2 antibodies to the
bacterium at different stages of natural infection (see Diagnosis,
later). The whole genome of C. burnetii has been sequenced.14
Etiology and Epidemiology C. burnetii is extremely contagious, and only small num-
Coxiella burnetii is a gram-negative, obligately intracellular, bers of organisms are required to cause disease in people. The
small, pleomorphic rod that causes Q fever in humans, a signifi- epidemiology of Q fever varies geographically. Because of its
cant zoonosis worldwide. Sheep, cattle, and goats are the most predilection for domestic ruminant species, Q fever is most com-
commonly reported reservoirs for the organism, but infected monly reported from rural areas. C. burnetii has been detected
cats and less commonly dogs have also been implicated in trans- within a huge variety of different tick and other arthropod spe-
mission to people.2-5 Animals other than humans are often sub- cies. Within ticks, it replicates in the midgut and it is shed in
clinically infected, although reproductive complications such as large quantities in tick feces. Although ticks have the potential
premature birth, abortion, and stillbirth may also occur. Res- to transmit the organism to domestic animal species, the most
ervoir hosts shed organisms in urine, feces, saliva, and/or milk, significant role of ticks appears to be in transmission to wild-
and especially in vaginal secretions and the products of partu- life.15 C. burnetii has been identified in fish, wild birds, rodents,
rition because of recrudescence of infection during pregnancy. marsupials, horses, swine, camels, marine mammals, ducks, tur-
The placentas of infected animals are heavily infected (up to keys, geese, cats, dogs, and rabbits. Although the mechanism is
109 organisms per gram of tissue). At parturition, aerosols are not clearly defined, cats and dogs may become infected through
created, which are subsequently inhaled by other animals and exposure to infected sheep, cattle, and goats—possibly through
humans (Figure 32-1). Ingestion of unpasteurized contaminated ingestion of unpasteurized milk from these species—and

320
CHAPTER 32  Coxiellosis and Q Fever 321

FIGURE 32-1  Transmission of Coxiella burnetii. The organism is most often transmitted to humans when animals give birth. People become infected by inhalation when they directly
contact the products of parturition or as a result of indirect contact with organisms that survive in the environment. Sheep, cattle, and goats are the most common reservoirs of the organ-
ism for human infection, but infected cats and less commonly dogs can also transmit the infection to humans. Ticks also harbor the organism and are thought to maintain transmission
to wildlife species, especially rodents (such as small mice) and rabbits, but also birds. Dogs and cats and humans can also become infected when they contact (or ingest) wildlife species.
Ingestion of infected milk may be a lesser mode of transmission.

through contact with, or predation of, infected wildlife reservoir organism appears to modulate apoptosis in order to inhibit
hosts such as rodents, birds, and rabbits. cell death and promote its own replication.23 Subsequently,
Exposure to stillborn kittens or apparently healthy newborn infected cells enter the bloodstream. After experimental infec-
kittens has been a risk factor for human infection in Africa, tion, bacteremia may be prolonged (up to 1 month) in cats.24
Japan, Korea, Canada, and the United States. In a study of intact The organism can persist in a variety of tissues, and in human
female cats from Colorado, C. burnetii DNA was not detected patients, this contributes to chronic disease manifestations, such
in vaginal swab specimens or uterine biopsies from 50 shelter as prolonged fever, endocarditis, vasculitis, chronic fatigue, and
cats using a PCR assay, but 4 of 47 uterine biopsies from client- immune-mediated consequences such as demyelinating poly-
owned cats were positive.16 Seroprevalence studies suggest that radiculoneuritis.25 Appropriate diagnosis and treatment at the
infection is more common in stray cats and dogs than client- acute stage of clinical illness is important in reducing the risk of
owned animals, but further research is required to determine postacute Q-fever sequelae and development of chronic disease.
risk factors for exposure in dogs and cats.17,18 No association Reactivation of shedding has been described during pregnancy
has been found between prevalence of Q fever in people and cat in humans.12 Infected cats and dogs usually show no clinical
or dog ownership.19 Laboratory-acquired infections have been signs of infection, but stillbirth, abortion, and prolonged vagi-
described in people, and infection can occur as a result of expo- nal bleeding for 3 weeks before delivery have been reported in
sure to straw, manure, or dust, such as that from contaminated cats.4,26 The organism replicates in trophoblasts, and in experi-
farm vehicles and contaminated clothing. Q fever has emerged mentally infected goats, massive bacterial replication occurs in
as a disease of U.S. combat arms military personnel who are the placenta just before abortion.27 An infected rabbit hound
deployed to Iraq.20 Studies of a large, 2-year outbreak in the dog that transmitted C. burnetii to a family in Nova Scotia
Netherlands showed that dust-accumulating surfaces contained gave birth to four puppies, all of which died within 24 hours
higher levels of C. burnetii DNA than vaginal swabs from sheep of birth.5 The importance of C. burnetii in reproductive tract
and goats.21 The organism can travel for miles as a result of disease and chronic disease manifestations such as endocarditis
windborne transmission. in cats and dogs requires further investigation.

Clinical Features Diagnosis


Signs and Their Pathogenesis Microbiologic Tests
After inhalation (and to a lesser extent, ingestion), C. burnetii Diagnostic assays available for Q fever are listed in Table 32-1.
enters mononuclear phagocytes and survives within the harsh
environment of the phagolysosome.22 Remarkably, the acid Culture
conditions of the lysosome appear to be a requirement for the C. burnetii can be isolated in a variety of different cell lines.
metabolic functions of C. burnetii. Replication is prolonged, It has also been isolated in a cell-free medium that mimics the
and minimal cytopathic effects occur in infected cells. The phagolysosome environment.28 Because it is highly infectious
322 SECTION 2  Bacterial Diseases

TABLE 32-1
Diagnostic Assays Available for Q Fever
Assay Specimen Type Target Performance
Cell culture Placental ­materials, C. burnetii Not widely offered or utilized for routine diagnostic purposes.
vaginal swabs, blood, Requires special facilities.
urine, feces, milk
Histopathology Placental materials C. burnetii Organisms may be seen when present in large numbers and stain
with Gimenez stain. Immunohistochemistry and fluorescence in
situ hybridization can be used to identify the organisms as C.
burnetii.
Serology Serum Antibodies to Acute and convalescent serology is required for diagnosis of acute
phase 1 or infection. At least 1 month may be required for seroconversion.
phase 2 Cross-reactivity may occur to related bacteria. IFA is considered
C. burnetii the serologic test of choice in human patients. CFT is the OIE
standard in animals but is less sensitive.
PCR Whole blood, tissue C. burnetii DNA Confirms active infection. Sensitivity and specificity may vary de-
specimens, vaginal pending on stage of infection, assay design, and specimen type.
swabs, milk, feces PCR is most sensitive in acute infections (in the first 2 weeks
of illness in humans), but is usually negative in humans with
chronic infections. Subclinical shedding in animals is variable
but is greatest during parturition.

CFT, Complement fixation test; IFA, immunofluorescent antibody; OIE, World Organization for Animal Health.

to humans, most laboratories do not have the special facilities test of choice for organism detection and have revolutionized
required to isolate C. burnetii. Thus diagnosis of infection is usu- knowledge of the epidemiology of C. burnetii infections. In ani-
ally performed using immunohistochemistry, serology, or PCR. mals, PCR assays have primarily been used on a research basis.
Negative results can occur with PCR assays in chronic infec-
Serologic Diagnosis tions or even as early as 2 weeks after onset of acute illness
The World Organization for Animal Health (OIE) reference test in humans, because of undetectable levels of the infection or
for serologic diagnosis of C. burnetii infection in animals remains absence of bacteremia. Because C. burnetii is highly infectious
the complement fixation test (CFT),20 despite the availability of to humans, special handling (BSL 3) conditions are required for
more sensitive serologic tests. A variety of serologic assays have fresh specimens that are submitted for C. burnetii PCR assays
been used to detect antibodies to phase 1 or phase 2 C. burnetii (see Table 1-4). When C. burnetii is suspected (such as in pla-
antigens in humans and other animals, including immunofluo- centas or aborted material), the laboratory should be contacted
rescent antibody (IFA) assays, ELISA, and the CFT. The CFT in advance, and specimens should be labeled and transported
is considered the least sensitive and specific method. In human appropriately.
patients, anti–phase 2 antibodies (IgG and IgM) are present in
acute Q fever, whereas anti–phase 1 IgG antibodies predominate Pathologic Findings
in chronic infection.15,29 In humans, the use of IFA assays has Little information is available about the pathology of C. bur-
been recommended for diagnosis of Q fever.12 Acute and con- netii infection in dogs and cats. In affected livestock, C. burnetii
valescent serology is required for diagnosis of acute Q fever in causes a necrotizing placentitis with mixed inflammatory cell
people. It can take at least a month before seroconversion occurs; infiltrate and nonsuppurative vasculitis. In some cases, small,
titers peak around 8 weeks after infection.15 Because infection is basophilic intracytoplasmic organisms can be seen (Figure 32-2).
usually not clinically detected in dogs and cats, serologic assays The organisms stain with Gimenez, Giemsa, and modified acid-
have primarily been used in dogs and cats for epidemiologic stud- fast stains, but not Gram, silver, or periodic acid–Schiff stains.32
ies or as part of import requirements to Q fever–free countries Immunohistochemistry and fluorescent in situ hybridization33
such as New Zealand. Low-level cross-reactivity with other bac- can be used to identify C. burnetii antigen within lesions.
teria such as Bartonella spp. and chlamydiae has been described
in some studies, which highlights the need for animal species- Treatment
specific cutoff values for all serologic methods.30,31 The concur-
rent performance of serology for Bartonella and Chlamydia (i.e., Many acute infections in people are self-limiting (or even sub-
to determine if titers are higher or lower to these organisms than clinical), and symptoms respond to nonspecific therapy such
to Coxiella) may assist in interpretation of the results. as antipyretics and rehydration. However, because of concerns
that relate to the development of chronic Q fever, specific anti-
Molecular Diagnosis Using the Polymerase Chain Reaction microbial therapy is recommended regardless of the severity
Both conventional and real-time PCR assays have been described of acute Q fever. When antimicrobials agents are required,
for detection of C. burnetii DNA. PCR assays are the diagnostic doxycycline for 14 to 21 days, azithromycin for 3 to 5 days,
CHAPTER 32  Coxiellosis and Q Fever 323

Prevention each year, and rates of hospitalization for these cases


were around 50%.37 However, severe cases are more likely to be
reported, so actual hospitalization rates are likely much lower.
In Australia, 300 to 400 cases are reported annually.38
In humans, acute Q fever occurs after an incubation period
that ranges from 9 to 39 days, although shorter incubation peri-
ods are possible with exposure to high doses of the agent.12 The
incubation period after exposure to an infected cat has ranged
from 9 days to 4 weeks.3,4 Males are predisposed, which relates
to an increased likelihood of exposure. Patients who develop
pneumonia may also develop severe headache, as well as sweats,
chills, fatigue, and myalgia. Other signs include cough, chest
pain, nausea and vomiting, sore throat, diarrhea, a rash, and
abdominal pain and hematuria.4 Myocarditis and pericardi-
tis can occur rarely. Blood work often shows a normal white
cell count, and thrombocytopenia or marked thrombocytosis
FIGURE 32-2  Histopathology showing severe, acute necrotizing placentitis can occur. Serum biochemistry testing reveals mild to moder-
in the placenta of a goat from which dead fetuses were removed by caesarian sec-
ate increases in the activity of hepatic transaminases in almost
tion. Trophoblasts are distended with abundant basophilic bacterial organisms
(arrows). The organisms were subsequently identified as Coxiella burnetii with all patients.12 Pulmonary infiltrates are present on thoracic
immunohistochemistry. radiographs, and in some cases, pleural effusion.12,15 Q fever
pneumonia is rarely fatal. Chronic Q fever may be manifested
as endocarditis, optic neuritis, osteomyelitis, granulomatous
or quinolones have been used successfully to treat pneumo- hepatitis, interstitial pulmonary fibrosis, prolonged fever, and
nia and hepatitis in human patients with C. burnetii infection. a chronic fatigue syndrome, which has been described in up to
Combination therapy, such as doxycycline, ciprofloxacin, and 20% of patients who develop acute infection with C. burnetii.39
rifampin for prolonged periods (e.g., 2 years) or doxycycline Endocarditis follows acute Q fever in fewer than 1% of cases,29
and hydroxychloroquine (which alkalinizes phagolysosomes) and it may not become apparent for several years after acute
has been required for treatment of Q fever endocarditis, and illness. Approximately 3% to 5% of human endocarditis cases
valve replacement is frequently needed.12,15 Antibody titers have been attributed to C. burnetii. Host factors seem to be
decrease with successful treatment. Q fever during pregnancy, important in whether chronic manifestations of Q fever occur.
which may result in abortion when infection occurs in the first Cellular immunity and the production of IFN-γ appear to be
trimester, is treated with trimethoprim-sulfamethoxazole for required for resolution of infection.29
the duration of the pregnancy.34 Treatment of an infected sheep Because treatment with tetracyclines does not seem to reduce
herd with oxytetracycline on days 100 and 120 of gestation did shedding by reservoir hosts, an emphasis has been placed on
not prevent shedding of C. burnetii in milk, vaginal swabs, or the development of effective vaccines for public health pur-
feces as assessed with PCR, nor did it affect the duration of poses. A phase 1 inactivated vaccine (Coxevac) has been used
shedding.35 in Europe on an investigational basis to prevent shedding by
reservoir livestock species, such as sheep and goats. Vaccina-
Public Health Aspects tion of sheep flocks led to a reduced number of abortions and
decreased shedding, but did not completely prevent infection.29
In humans, asymptomatic infections with C. burnetii are proba- Vaccination was ineffective in previously infected cattle or naïve
bly widespread. When it occurs, Q fever is most often a systemic pregnant cattle.40 Another phase 1 whole cell vaccine (Q-VAX,
febrile illness. Because it mimics a variety of other illnesses, and Commonwealth Serum Laboratories, Parkville, Australia) has
confirmatory diagnostic tests require a specific index of suspi- also been used to protect naïve but at-risk humans, such as
cion by the medical clinician, Q fever is most likely underrecog- livestock workers (farmers, shearers, slaughterhouse workers,
nized. Veterinarians involved in mixed or farm animal practice, animal transporters), veterinarians, and laboratory workers,
slaughterhouse workers, and workers in the livestock industry from infection. A Q-VAX skin test as well as serologic assays
(farmers, shearers, transporters of animals) are most likely to are performed as a prescreening of potential vaccine recipients
develop Q fever as a result of direct contact with infected live- for prior sensitization to Q fever antigens to minimize adverse
stock. Person-to-person spread of Q fever is rarely reported, but vaccine reactions; humans with prior exposure to C. burnetii
humans can shed organisms during parturition, and infection should never be vaccinated. It has been recommended that
of an obstetrician during delivery was described.36 Transmis- humans not drink raw milk from infected farms. When Q fever
sion during postmortem examinations has also been reported. is discovered on a farm, sale of milk is forbidden for the year
Between 2005 and 2008, approximately 140 to 170 cases of after initial diagnosis. Aborted material and placentas must be
Q fever were reported to the Centers for Disease Control and buried with lime or incinerated.29
324 SECTION 2  Bacterial Diseases

CASE EXAMPLE chest pain, nausea, and vomiting. As a result, she was hospi-
talized for 3 weeks for further diagnostic testing, treatment,
and convalescence. When the technician tested positive by
Signalments: 50-year-old female veterinarian and a 23-year- PCR assay for Coxiella burnetii DNA on day 10 after the onset
old female veterinary technician from southeastern Australia of illness, serial diagnostic tests for Q fever were also per-
History: A 3-year-old female British shorthair cat with dystocia formed on the veterinarian (Table 32-2).
was brought to a veterinary clinic by the breeder/owner Comments: Determination of the date of onset of illness is
at 8 o’clock in the morning. One kitten was stillborn in the essential in any diagnostic investigation of Q fever in people
home environment overnight. Safe delivery of the two for correct interpretation of diagnostic test results. In the first
remaining kittens required a cesarean section. Surgery was 8 to 10 days after the onset of illness, patients are frequently
performed immediately, and the kittens were delivered and (80%) bacteremic (and therefore PCR positive) but still
revived by vigorous rubbing with dry towels. The kittens antibody negative. IgM class antibody to phase 2 antigens,
were discolored by yellow-green placental material and as measured via IFA or ELISA assays, appear as early as 10 to
were slow to revive. They were able to successfully to suckle 12 days after the onset of illness, but this may also be
once the queen had recovered from general anesthesia. The delayed, as occurred in the veterinarian in this example. IgM
queen and kittens were returned to the breeder’s cattery the class antibody to phase 1 and IgG class antibody to phase
same afternoon. 2 antigens often appear on day 15 to 20, but again this
Two weeks after the surgery, the veterinarian and the vet- can be delayed. Small animal veterinarians and veterinary
erinary technician who had been involved in the cesarean technicians or nurses who are exposed to kittens and placental
section developed fever, severe headache, neck pain, chills, secretions and subsequently develop illness should inform
night sweats, myalgia, and stiffness. The veterinarian sought their practitioner of the possibility of exposure to C. burnetii
medical attention. Psittacosis was considered the most likely because diagnosis requires a high index of suspicion and
diagnosis by the attending physician, because psittacine specific diagnostic assays. Fatigue often persists in patients
birds were frequently treated in the clinic. A serologic test for months after the onset of illness (as was the case for
for chlamydiosis was negative, but nevertheless treatment the veterinary technician in this example) and does not
with doxycycline was commenced. The veterinary techni- necessarily mean that chronic Q fever has developed.
cian’s clinical signs were more severe and included cough,

TABLE 32-2
Results of Diagnostic Assays for Q fever in a Veterinarian and Veterinary Technician after Exposure to an Infected
Cat’s Reproductive Secretions

Real-Time PCR on Whole Serology for IgM and IgG to Phase 1 and Phase 2 Antigens*
Days after Blood
the Onset of
Illness Veterinarian Technician Veterinarian Technician
10 ND Positive ND Negative for IgG and IgM to phase
2 antigen
14 Negative ND Negative ND
20 Negative Negative Negative Positive for IgM to phase 1 and
phase 2 antigens, positive for IgG
to phase 2 antigen. Positive CFT
for antibodies to phase 2 antigen
35 ND ND Positive for IgM and IgG to ND
phase 2 antigen.
Positive CFT for antibodies to
phase 2 antigens

CFT, Complement fixation test; ND, not determined.


*Performed by immunofluorescent antibody unless otherwise specified.
CHAPTER 32  Coxiellosis and Q Fever 325

SUGGESTED READINGS 20. Anderson AD, Baker TR, Littrell AC, et al. Seroepidemiologic sur-
vey for Coxiella burnetii among hospitalized U.S. troops deployed
Angelakis E, Raoult D. Q Fever. Vet Microbiol. 2010;140:297-309. to Iraq. Zoonoses Public Health. 2011;58:276-283.
Porter SR, Czaplicki G, Mainil J, et al. Q Fever: current state of knowl- 21. de Bruin A, de Groot A, de Heer L, et  al. Detection of Coxiella
edge and perspectives of research of a neglected zoonosis. Int J Micro- burnetii in complex matrices by using multiplex quantitative PCR
biol. 2011;2011:248-418. during a major Q fever outbreak in The Netherlands. Appl Environ
Microbiol. 2011;77:6516-6523.
REFERENCES 22. Voth DE, Heinzen RA. Lounging in a lysosome: the intracellular
1. Derrick EH. “Q” fever, a new fever entity: clinical features, diagno- lifestyle of Coxiella burnetii. Cell Microbiol. 2007;9:829-840.
sis and laboratory investigation. Med J Aust. 1937;2:281-299. 23. Voth DE, Howe D, Heinzen RA. Coxiella burnetii inhibits apop-
2. Marrie TJ, Langille D, Papukna V, et al. Truckin’ pneumonia—an tosis in human THP-1 cells and monkey primary alveolar macro-
outbreak of Q fever in a truck repair plant probably due to aero- phages. Infect Immun. 2007;75:4263-4271.
sols from clothing contaminated by contact with newborn kittens. 24. Gillespie JH, Baker JA. Experimental Q fever in cats. Am J Vet Res.
Epidemiol Infect. 1989;102:119-127. 1952;13:91-94.
3. Kosatsky T. Household outbreak of Q-fever pneumonia related to 25. Lefebvre M, Grossi O, Agard C, et  al. Systemic immune presen-
a parturient cat. Lancet. 1984;2:1447-1449. tations of Coxiella burnetii infection (Q fever). Semin Arthritis
4. Marrie TJ, MacDonald A, Durant H, et  al. An outbreak of Rheum. 2010;39:405-409.
Q fever probably due to contact with a parturient cat. Chest. 26. Nagaoka H, Sugieda M, Akiyama M, et  al. Isolation of Coxiella
1988;93:98-103. burnetii from the vagina of feline clients at veterinary clinics. J Vet
5. Buhariwalla F, Cann B, Marrie TJ. A dog-related outbreak of Q Med Sci. 1998;60:251-252.
fever. Clin Infect Dis. 1996;23:753-755. 27. Sanchez J, Souriau A, Buendia AJ, et al. Experimental Coxiella bur-
6. Fishbein DB, Raoult D. A cluster of Coxiella burnetii infections netii infection in pregnant goats: a histopathological and immuno-
associated with exposure to vaccinated goats and their unpasteur- histochemical study. J Comp Pathol. 2006;135:108-115.
ized dairy products. Am J Trop Med Hyg. 1992;47:35-40. 28. Omsland A, Cockrell DC, Howe D, et al. Host cell-free growth of
7. Benson WW, Brock DW, Mather J. Serologic analysis of a peni- the Q fever bacterium Coxiella burnetii. Proc Natl Acad Sci U S A.
tentiary group using raw milk from a Q fever infected herd. Public 2009;106:4430-4434.
Health Rep. 1963;78:707-710. 29. Porter SR, Czaplicki G, Mainil J, et  al. Q Fever: current state of
8. Raoult D, Marrie T, Mege J. Natural history and pathophysiology knowledge and perspectives of research of a neglected zoonosis. Int
of Q fever. Lancet Infect Dis. 2005;5:219-226. J Microbiol. 2011;2011:248-418.
9. Vapniarsky N, Barr BC, Murphy B. Systemic Coxiella-like infec- 30. La Scola B, Raoult D. Serological cross-reactions between Barton-
tion with myocarditis and hepatitis in an eclectus parrot (Eclectus ella quintana, Bartonella henselae, and Coxiella burnetii. J Clin
roratus). Vet Pathol. 2012;49(4):717-722. Microbiol. 1996;34:2270-2274.
10. Shivaprasad HL, Cadenas MB, Diab SS, et al. Coxiella-like infec- 31. Lukacova M, Melnicakova J, Kazar J. Cross-reactivity between
tion in psittacines and a toucan. Avian Dis. 2008;52:426-432. Coxiella burnetii and chlamydiae. Folia Microbiol (Praha).
11. Woc-Colburn AM, Gamer MM, Bradway D, et al. Fatal coxiello- 1999;44:579-584.
sis in Swainson’s Blue Mountain rainbow lorikeets (Trichoglossus 32. Moore JD, Barr BC, Daft BM, et al. Pathology and diagnosis of Cox-
haematodus moluccanus). Vet Pathol. 2008;45:247-254. iella burnetii infection in a goat herd. Vet Pathol. 1991;28:81-84.
12. Marrie TJ, Raoult D. Coxiella burnetii (Q fever). In: Mandell GL, 33. Jensen TK, Montgomery DL, Jaeger PT, et al. Application of fluo-
Bennett JE, Dolin R, eds. Mandell, Douglas and Bennett’s Prin- rescent in situ hybridisation for demonstration of Coxiella burnetii
ciples and Practice of Infectious Diseases. 7th ed. Philadelphia, PA: in placentas from ruminant abortions. APMIS. 2007;115:347-353.
Churchill Livingstone Elsevier; 2011:2511-2519. 34. Carcopino X, Raoult D, Bretelle F, et al. Managing Q fever during
13. Jerrells TR, Hinrichs DJ, Mallavia LP. Cell envelope analy- pregnancy: the benefits of long-term cotrimoxazole therapy. Clin
sis of Coxiella burneti phase I and phase II.. Can J Microbiol. Infect Dis. 2007;45:548-555.
1974;20:1465-1470. 35. Astobiza I, Barandika JF, Hurtado A, et  al. Kinetics of Coxiella
14. Seshadri R, Paulsen IT, Eisen JA, et al. Complete genome sequence burnetii excretion in a commercial dairy sheep flock after treatment
of the Q-fever pathogen Coxiella burnetii. Proc Natl Acad Sci U S A. with oxytetracycline. Vet J. 2010;184:172-175.
2003;100:5455-5460. 36. Racult D, Stein A. Q fever during pregnancy—a risk for women,
15. Angelakis E, Raoult D. Q Fever. Vet Microbiol. 2010;140:297-309. fetuses, and obstetricians. N Engl J Med. 1994;330:371.
16. Cairns K, Brewer M, Lappin MR. Prevalence of Coxiella burnetii 37. Centers for Disease Control and Prevention. http://www.cdc.gov/
DNA in vaginal and uterine samples from healthy cats of north- qfever/stats/index.html. Last accessed December 13, 2012.
central Colorado. J Feline Med Surg. 2007;9:196-201. 38. Australian Government Department of Health and Aging.
17. Komiya T, Sadamasu K, Kang MI, et al. Seroprevalence of Coxiella http://www.health.gov.au/internet/publications/publishing.nsf/Con
burnetii infections among cats in different living environments. tent/cda-cdi34suppl.htm. Last accessed December 13, 2012.
J Vet Med Sci. 2003;65:1047-1048. 39. Marmion BP, Shannon M, Maddocks I, et  al. Protracted debility
18. Willeberg P, Ruppanner R, Behymer DE, et  al. Environmental and fatigue after acute Q fever. Lancet. 1996;347:977-978.
exposure to Coxiella burnetii: a sero-epidemiologic survey among 40. Guatteo R, Seegers H, Joly A, et al. Prevention of Coxiella burnetii
domestic animals. Am J Epidemiol. 1980;111:437-443. shedding in infected dairy herds using a phase I C. burnetii inacti-
19. Skerget M, Wenisch C, Daxboeck F, et al. Cat or dog ownership vated vaccine. Vaccine. 2008;26:4320-4328.
and seroprevalence of ehrlichiosis, Q fever, and cat-scratch disease.
Emerg Infect Dis. 2003;9:1337-1340.
CHAPTER 33

Chlamydial Infections
Jane E. Sykes

room temperature and they are readily inactivated by most dis-


Overview of Chlamydial Infections in Dogs infectants. Transmission occurs via direct contact or, to a lesser
and Cats extent, aerosols. Fomites are also likely to be an important
First Described in Cats: 1944, United States (Baker)1 means of transmission among group-housed cats in heavily con-
Causes: Chlamydia felis (and possibly other chlamydiae such taminated environments. The chlamydiae at one time were sepa-
as Chlamydia pneumoniae) causes ocular disease in cats; rated into two genera, Chlamydia and Chlamydophila, based
Chlamydia psittaci and Chlamydia abortus have rarely on sequence analysis of the 16S rRNA and 23S rRNA genes.
been detected in association with ocular and respiratory However, there has been disagreement in the field regarding this
disease in dogs. These are obligately intracellular bacteria classification, and reversion to the single genus Chlamydia has
that belong to the family Chlamydiaceae. since been recommended based on the availability of complete
genome sequence information.2
Geographic Distribution: Worldwide
Chlamydiae persistently infect epithelial cells of the ocular,
Mode of Transmission: Direct contact with respiratory secre- respiratory, gastrointestinal, and genitourinary systems; chla-
tions and possibly also vaginal secretions and feces mydial infections of the synovium have also been reported in
Major Clinical Signs (Cats): Acute and chronic conjunctivi- a number of different host species. Different chlamydial species
tis, chemosis, serous to mucopurulent ocular discharge, tend to associate with certain host species, although evidence
and blepharospasm, with or without sneezing and nasal has accumulated that some chlamydial species can cause disease
discharge in more than one host species (Table 33-1). In addition, more
than one chlamydial species can cause similar disease manifesta-
Differential Diagnoses: The primary differential diagnoses in
tions in a single host species. For example, although the human
cats are infections with feline herpesvirus 1, feline calici-
ocular disease trachoma is caused primarily by C. trachomatis,
virus, Mycoplasma spp., and/or Bordetella bronchiseptica
it has now also been associated with infection by C. pneumoniae
Human Health Significance: C. felis has rarely been docu- and C. psittaci.3 Chlamydia felis is an important cause of con-
mented as a cause of human conjunctivitis. The impor- junctivitis and possibly reproductive disease in cats, but DNA
tance of cats and dogs as a source of other human that resembles that of the human pathogen C. pneumoniae has
chlamydial infections is unclear. C. psittaci is generally now also been detected in ocular swabs from European cats with
acquired from avian species and not dogs and cats. conjunctivitis.4 Chlamydia abortus DNA was detected in histo-
pathology specimens from 1 of 13 cats with arteriosclerosis.5
There are rare reports of chlamydial infections in dogs, in asso-
ciation with ocular and respiratory disease, polyarthritis, and
Etiology Agent and Epidemiology atherosclerosis.6-11 However, chlamydiae were not detected by
PCR assay in atherosclerotic lesions from 16 dogs in one study,
Chlamydiae are obligately intracellular bacteria that belong to so the role of chlamydiae in this otherwise rare condition in dogs
the order Chlamydiales. They primarily cause conjunctivitis in is uncertain.5 Respiratory disease associated with C. psittaci
cats, but may also play a role in other systemic and reproduc- infection has been reported most frequently among dogs.
tive disorders in dogs and cats that remain poorly character- The most significant pathogen of cats is C. felis, which causes
ized. Most knowledge of chlamydial infections results from acute and chronic conjunctivitis in cats worldwide. The results of
research on human chlamydial pathogens such as Chlamydia genetic analysis (by multilocus variable-number tandem-repeat
trachomatis, Chlamydia pneumoniae, and Chlamydia psittaci, and multilocus sequence typing) have revealed that as many as
which have significant clinical and public health importance (see 25 different genotypes of C. felis exist, and more than one geno-
Public Health Significance). Chlamydiae structurally resemble type may circulate in some catteries.12 Co-infections with more
gram-negative bacteria, lack peptidoglycan, and have extremely than one strain can occur. The prevalence of infection varies
small genomes; they depend on the host cell for many amino from one group of cats to another and with geographic location,
acids and nucleotides. They exist in two forms, an infectious age of the cat population sampled, and presence or absence of
elementary body (EB), which is 0.2 to 0.6 µm in diameter and clinical illness. Infection occurs most often in ­multiple-cat house-
exists outside the cell, and the reticulate body (RB), which is holds, especially breeding catteries. C. felis DNA is uncommonly
larger (0.5 to 1.5 µm), is noninfectious, and replicates within the detected in conjunctival swabs from healthy cats. For example,
cytoplasm of host cells, before it matures and forms EBs (Figure in clinically healthy household cats, those with a history of past
33-1). The EBs survive only a few days in the environment at conjunctivitis, and those with active conjunctivitis, 0%, 4.6%,

326
CHAPTER 33  Chlamydial Infections 327

Attachment
Phagosome fusion and 7.3% had positive PCR assay results, respectively.13 Isola-
Reorganization tion rates of up to 30% in household cats with conjunctivitis
and ingestion RB EB to RB
have been reported.14 In shelter cats, the prevalence of C. felis
EB 2hr 8hr
infection has ranged from 0% to 15%.15-18 In cats from 218
8-18hr Multiplication European rescue shelters, breeding establishments, and private
AB of RB households, the prevalence of infection in cats without respi-
ratory disease was 3%, versus 10% in those with evidence of
respiratory disease.19 Infection with C. felis in this European
Persistence
18-24hr study was associated with suboptimal hygiene. Infection with
C. felis is more often present in young cats, especially those aged
Extrusion and 2 to 12 months; kittens less than 2 months of age may be pro-
release of 48-72hr Condensation tected by maternal antibody, although neonatal infections have
infectious EBs 24-48hr RB to EB been described. Cats older than 5 years of age are very unlikely
A Mature inclusion to be infected with C. felis. Co-infections with other respiratory
pathogens, such as feline herpesvirus 1 (FHV-1) and feline cali-
civirus (FCV), are common.
The DNA of organisms that belong to the family Parachla-
mydiaceae has recently been detected in cats with ocular disease.
These organisms resemble Chlamydia and reside symbiotically
within amoebae. Parachlamydial species include ­Neochlamydia,
Parachlamydia, Protochlamydia, ­Rhabdochlamydia, ­Criblamydia,
Simkania, and Waddlia species. DNA that resembles that of
­Neochlamydia hartmanellae has been detected in feline conjunc-
tival brush specimens.20 This organism is an endosymbiont of the
amoeba Hartmannella vermiformis, which has been identified as
B a cause of ocular surface infection in people and may play some
role in feline ocular infections. The DNA of Parachlamydia acan-
FIGURE 33-1  A, Life cycle of chlamydiae. From top left. Electron-dense elementary thamoebae has also been detected with PCR in cats with keratitis
bodies (EBs) attach to and are taken up by epithelial cells. EBs then differentiate into and conjunctivitis.21 As yet, co-infections with amoebae have not
reticulate bodies (RBs), and RBs divide by binary fission. RBs condense into EBs, and also been detected in cats. Further studies are required to determine the
have the potential to persist in the form of aberrant bodies (AB). EBs are released by cell
prevalence and clinical significance of these organisms.
lysis and infect other cells. B, Electron micrograph of chlamydial organisms growing in an
inclusion in tissue culture. The larger reticulate bodies (RBs) have more diffuse chromatin.
One of the RBs appears to be dividing. The smaller dense bodies are elementary bodies.   Clinical Features
(B from Mandell GL, Bennett JE, Dolin R. Principles and Practice of Infectious Diseases. Ed
7. Philadelphia, PA: Elsevier, 2012; 2:2440) Signs and Their Pathogenesis
C. felis infection is acquired by cats primarily through close
contact, fomites, or to a lesser extent, aerosol transmission. Kit-
tens may be infected from the mother at birth. Whether venereal

TABLE 33-1
Chlamydial Species That Infect Animals and Humans
Species Major Host(s)* Major Clinical Manifestations
Chlamydia trachomatis Humans Conjunctivitis (serovars A-C); urethritis, ­cervicitis,
proctitis (serovars D-K); lymphogranuloma
­venereum (serovars L1, L2, and L3)
Chlamydia pneumoniae Humans Upper respiratory infection, community-acquired
pneumonia, possibly ­atherosclerosis and asthma
Chlamydia psittaci Humans, a variety of bird species (e.g., parrots, Pneumonia, systemic ­infection
finches, poultry, pigeons, pheasants, seagulls,
egrets, puffins)
Chlamydia felis Cats Conjunctivitis, upper respiratory infection, arthritis?,
reproductive disease?
Chlamydia abortus Sheep, cows, goats, other mammals Reproductive disease
Chlamydia pecorum Cattle and sheep Encephalomyelitis, pneumonia, reproductive tract
disease, conjunctivitis, polyarthritis
Chlamydia caviae Guinea pigs Keratoconjunctivitis

*Infection may occasionally extend to other host species.


328 SECTION 2  Bacterial Diseases

transmission occurs, as reported for other chlamydial species,


is unknown, but in some infected cats, the organism is shed
in vaginal discharges and from the rectum as well as in ocu-
lar discharges. After uptake of EBs, the EB transitions into the
RB, which replicates by binary fission in a membrane-enclosed
vacuole called an inclusion and avoids lysosomal fusion. The
RBs then transition back to the EB form, which is released into
the extracellular milieu after cell lysis occurs and subsequently
infect other host cells. The entire replication cycle takes about
2 days to complete (see Figure 33-1).22
Chlamydiae primarily infect epithelial cells, but they can
also infect a variety of other cell types including endothelial
cells, smooth muscle cells, lymphocytes, monocytes, and mac-
rophages. C. felis replicates in the cytoplasm of conjunctival
epithelial cells, but also spreads via the bloodstream to a variety
of other tissues, such as the tonsil, lung, liver, spleen, gastroin-
testinal tract, and kidney.23 Infection is typically followed 2 to
5 days later by the development of acute, chronic, or recurrent FIGURE 33-2  Domestic shorthair cat with acute chlamydial conjunctivitis.
conjunctivitis, with or without signs of rhinitis such as nasal dis- Marked hyperemia and chemosis are present, as well as serous to mucopurulent ocular
charge and sneezing. Most cats remain otherwise well and appe- discharge.
tent. Lower respiratory tract signs such as cough and dyspnea
rarely, if ever, occur. Corneal disease such as keratitis or corneal
ulceration is rare and, if present, is likely caused by other or involvement, such as stertorous respiration, serous or mucopu-
co-infecting pathogens such as FHV-1. Experimental inocula- rulent nasal discharge, and sneezing, may accompany conjuncti-
tions have also been followed by signs of fever, lethargy, lame- vitis. Mucopurulent vaginal discharge has been reported in some
ness and joint pain (possibly due to polyarthritis), and weight cats that were experimentally infected by topical ophthalmic
loss, in addition to conjunctivitis.24 There are also rare reports inoculation.
of gastritis and peritonitis in association with the detection of Physical examination findings described in dogs with chla-
chlamydia-like organisms in cats.25,26 mydiosis have included fever, tachypnea, keratoconjunctivitis
After the initial episode of acute conjunctivitis, infection may with mucopurulent ocular discharge, shifting leg lameness, and
persist for many months. This may be accompanied by mild lymphadenopathy.6-9,11
conjunctivitis or, in some cases, no clinical signs of disease.
Organisms can be isolated from the conjunctiva for up to 215 Diagnosis
days after experimental infection.27,28 Whether chronic disease
is the result of repeated reinfection or the presence of persis- There are no clinically significant laboratory abnormalities in
tent chlamydiae is unclear. Co-infections with other agents such cats with chlamydiosis, and there is insufficient information
as FCV, FHV-1, Bordetella, or Mycoplasma may increase the on laboratory abnormalities that might occur in affected dogs.
severity of disease and duration of shedding. Other bacteria, Diagnosis of chlamydiosis is most often based on the results of
including those that normally colonize the healthy conjunctival molecular testing using PCR assays (Table 33-2). For all diag-
sac, also act as secondary invaders and worsen disease. nostic tests that detect chlamydial organisms, sufficient num-
Whether C. felis causes reproductive disease in cats remains bers of infected epithelial cells must be collected. This generally
unclear. The stage of pregnancy at which infection occurs, involves vigorous swabbing of infected mucosal sites such as the
co-infection with other organisms such as FHV-1, concurrent conjunctiva.
immunosuppression, route of infection, and the strain involved
may be important in determining the ability of C. felis to cause Microbiologic Tests
reproductive dysfunction, if this indeed occurs in cats. Cytologic Examination for Inclusions
Chlamydial infections are infrequently described in dogs. Using light microscopy, chlamydial intracytoplasmic inclusions
Chlamydia psittaci has been detected in dogs with respiratory may be seen in epithelial cells that are present in smears of con-
disease, such as pneumonia and pleural effusion, and/or kera- junctival scrapings. Inclusions are made up of basophilic clus-
toconjunctivitis.6-8 One group of bitches that had ocular and ters of coccoid bacteria and are up to 10 µm in diameter (Figure
lower respiratory signs as well as stillbirths and low litter size 33-3). Giemsa is the preferred staining method. Inclusions are
had nasal, conjunctival, and pharyngeal swab specimens that only generally visible early, if at all, during the course of infec-
tested positive for C. psittaci with a PCR assay.6 Chlamydia tion, so this method is insensitive. Also, melanin granules in the
abortus was detected in a dog with keratoconjunctivitis.9 The cytoplasm of conjunctival epithelial cells can yield false-positive
DNA of C. pneumoniae was detected in atherosclerotic lesions results. The use of direct fluorescent antibody staining or immu-
from dogs in Japan,10 and chlamydia-like organisms were found nocytochemical stains that detect Chlamydia increases sensitiv-
in a dog from South Africa with polyarthritis.11 ity and specificity.

Physical Examination Findings Enzyme-Linked Immunosorbent Assay Antigen Detection


Physical examination findings in cats with chlamydiosis A variety of commercial antigen detection enzyme immunoassay
include conjunctivitis, chemosis, serous to mucopurulent ocu- kits have been developed and marketed for diagnosis of human
lar discharge, and blepharospasm (Figure 33-2). Signs of nasal chlamydial infections. Nucleic acid amplification tests have now
CHAPTER 33  Chlamydial Infections 329

TABLE 33-2
Diagnostic Assays Available for Chlamydiosis in Dogs and Cats
Assay Specimen Type Target Performance
Cell culture Conjunctival swabs Chlamydia spp. Not widely offered or utilized for routine diagnostic pur-
poses. Requires special chlamydial transport media. False
negatives occur when organisms lose viability during
transport or in chronic infections, when organism numbers
are very low.
Cytology Conjunctival smears Chlamydial inclusions Low sensitivity. Inclusions are most likely to be seen early
in infection. False positives can occur when cytoplasmic
granules are mistaken for inclusions. Fluorescent or immu-
noperoxidase antibody techniques may increase sensitivity
and specificity.
Serology Serum Antibodies to Acute and convalescent serology is required for diagnosis of
Chlamydia spp. acute infection. Vaccination may interfere with interpreta-
tion of results. Not widely available or utilized worldwide.
PCR Conjunctival swabs, Chlamydia felis DNA or Confirms active infection. Assays may be genus or species
scrapings, or biopsies Chlamydia spp. DNA specific. Sensitivity and specificity may vary depending on
assay design and specimen type. Because healthy animals
occasionally test PCR positive, positive PCR results must
be interpreted in light of the clinical signs. Vaccination with
attenuated live chlamydial vaccines has the potential to
interfere with results.

antigen have lower sensitivity and specificity than cell culture,


direct fluorescent antibody methods, or PCR assays for detec-
tion of C. felis.

Cell Culture
Cell culture has traditionally been considered the gold standard
for chlamydial diagnosis, but is now mostly performed on a
research basis. Isolations from cats are usually highest from con-
junctival swabs, although nasal and pharyngeal swab specimens
may also contain infectious organisms. To enhance organism
survival, swabs should be placed immediately in a chlamydial
transport medium such as 2SP (0.2 M sucrose and 0.02 M phos-
phate), which can be obtained from laboratories that perform
chlamydial culture. Routine viral transport media should not be
used, because it contains antibiotics that inactivate the organisms.
The specimen should be refrigerated (4°C) if it is not immediately
sent to the laboratory and ideally should reach the laboratory
within 24 hours of specimen collection. In the laboratory, organ-
isms are readily grown in the yolk sacs of embryonating eggs or
FIGURE 33-3  Conjunctival scraping from a cat with chlamydial conjunctivitis. in cell lines such as McCoy or HeLa cells over a period of 2 to
Inclusions of Chlamydia felis are found in an epithelial cell (arrows). Wright’s stain, original 3 days. Fluorescent antibody is then used to identify chlamydial
magnification 1000×. (Courtesy of Judith Taylor, LabVet Consultations, Inc., Guelph, Ontario, inclusions within the infected cell culture (Figure 33-4).
Canada and Karen M. Young, University of  Wisconsin-Madison. In Young KM, Taylor JJ: Labora-
tory medicine: yesterday today tomorrow: eye on the cytoplasm, Vet Clin Pathol 35:141, 2006.) Molecular Diagnosis Using the Polymerase Chain Reaction
The use of PCR-based assays has revolutionized diagnosis of
chlamydial infections in animals and humans, because these
largely replaced the use of these assays for diagnosis of human assays are rapid, sensitive, and specific and do not require spe-
infections. Antibodies used in ELISA assays that are designed to cial transport conditions. Sequence analysis of PCR products
detect human chlamydial pathogens in swab specimens (such as can be used to differentiate between species of Chlamydiaceae.
conjunctival swabs) cross-react with C. felis antigen and so have Both conventional and real-time PCR assays have been used to
also been used for diagnosis of feline chlamydiosis. Unfortu- detect C. felis in conjunctival swab specimens from cats,29-31 and
nately, the kits have varied considerably in their sensitivity and real-time PCR assays that detect C. felis are now offered by sev-
specificity. In general, ELISA methods that detect chlamydial eral commercial veterinary diagnostic laboratories worldwide.
330 SECTION 2  Bacterial Diseases

TABLE 33-3
Suggested Medications for Treatment of Cats
with Ocular Chlamydiosis
Interval
Drug Dose Route (hours) Duration
Doxycycline* 10 mg/kg PO 24 At least 4 weeks,
or 2 weeks after
resolution of
­clinical signs
Amoxicillin- 12.5 PO 12 4 weeks
clavulanate
FIGURE 33-4  Chlamydial inclusions stained with fluorescent antibody in a mccoy cell Pradofloxacin 5-7.5† PO 24 6 weeks
monolayer. (Courtesy of Robert Suchland, Seattle, In Stamm WE, Batteiger BE. Introduc-
tion to Chlamydia and Chlamydophila. In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, *Doxycycline is the treatment of choice. For cats that fail to tolerate
Douglas and Bennett’s Principles and Practice of Infectious Diseases, 7 ed. Philadelphia, doxycycline, amoxicillin-clavulanate should be used. Fluoroquinolones
PA: Churchill Livingstone Elsevier; 2011.) are an option for cats that do not tolerate doxycycline or amoxicillin-
clavulanate.
†Dose is for the oral suspension; if tablets are used, the dose is lower

(3 mg/kg).
Unfortunately, considerable interlaboratory variation in assay
performance still exists.32 In addition, PCR assays for C. felis
may not detect other chlamydial species, such as C. pneumoniae. Treatment and Prognosis
Genus- or family-specific chlamydial PCR assays must be used
to detect these organisms. The laboratory should be consulted Chlamydial infections are generally treated with tetracycline
to determine the specificity of the assay used. The use of a DNA antibiotics. Oral administration of doxycycline at 5 to 10
microarray assay for detection and identification of chlamydiae mg/kg q12h for 3 to 4 weeks results in clinical resolution in
in cats has also been reported.5 The microarray assay consists of most cats. A dose of 10 mg/kg q24h can also be used (Table
a chip that carries chlamydial species- and genus-specific probes. 33-3).23 Clinical improvement occurs within 24 to 48 hours
DNA within the specimen is first amplified and biotin labeled after treatment is initiated. In colonies of cats, all cats should
with PCR, and then reacted with the microarray probes.33 be treated for 4 weeks: in one study that used real-time PCR
PCR assays for C. felis are usually more sensitive compared to detect infection, a 3-week course of treatment was insuf-
with cell culture for diagnosis of infection in naturally infected ficient to clear infection in some cats, but treatment for 28
cats (see Table 33-1).29 During antimicrobial treatment of days subsequently cleared infection in all cats.30 Treatment
infected cats, PCR assay results continue to be positive for sev- for up to 6 to 8 weeks may be required. Regardless, treatment
eral days.29,30 Topical ophthalmic solutions such as proxymeta- should continue for at least 2 weeks after the resolution of
caine and fluorescein did not interfere with the ability of clinical signs.
real-time PCR to detect C. felis, but there was a slight inhibitory Failure to respond completely to treatment may reflect the
effect when fusidic acid was used.34 Because cats without signs concurrent presence of viral upper respiratory tract pathogens.
of conjunctivitis occasionally test PCR positive for C. felis, the Because administration of doxycycline hyclate can result in
results of PCR must be interpreted in light of the history, clinical esophagitis and esophageal stricture formation, the administra-
signs present, and response to appropriate treatment. tion of doxycycline hyclate tablets should be followed by a bolus
of water or food (see Chapter 8 for additional information on
Serologic Testing doxycycline). Ocular infections may initially respond to tetracy-
Serologic tests are not widely available for routine diagnosis cline eye ointment applied two or three times daily,37 but recur-
of chlamydial infections. Assays that detect serum antibod- rent infections have been documented,38 likely because systemic
ies to chlamydiae, such as IFA or ELISA assays, are of limited infection with chlamydiae can occur in cats with conjunctivitis.
benefit for detection of active infection because the IgG anti- For cats that do not tolerate doxycycline, 4 weeks of
body titer increase is variable or prolonged, and IgM antibody amoxicillin-clavulanate is a suitable alternative.39 In exper-
titers are inconsistently increased. However, very high anti- imentally infected cats, the efficacy of enrofloxacin (5 mg/
body titers (≥1:512) have been associated with clinical illness kg PO q24h for 14 days) was similar to that of doxycycline
and/or chlamydial shedding, and high antibody titers among as determined with direct fluorescent antibody on conjunc-
group-housed cats with conjunctivitis can suggest a role for tival swabs, although a few cats in both the doxycycline and
C. felis in disease.13,23,35 Chlamydial infections persist in the enrofloxacin groups had evidence of persistent infection at
presence of high serum antibody titers, so positive antibody the end of the treatment period.40 Doxycycline is preferred
titers after vaccination do not imply protection. Serial titers may because of concerns that relate to the possibility of reti-
be needed to document active infections, and recent vaccination nal toxicity in cats treated with enrofloxacin. Doxycycline
may interfere with interpretation of positive titer results. An was more efficacious than pradofloxacin in experimentally
ELISA assay that discriminates between the antibody response infected cats that were monitored with a PCR assay.41 Sul-
from vaccination and that from natural infection has been fonamides or chloramphenicol are ineffective, and in  vitro,
described but is not widely available on a commercial basis.36 penicillin is only inhibitory at higher doses. Treatment with
CHAPTER 33  Chlamydial Infections 331

azithromycin is also incompletely effective when compared Public Health Aspects


with doxycycline.42
The most significant chlamydial pathogens of humans are C.
Immunity and Vaccination trachomatis, C. pneumoniae, and C. psittaci. C. trachoma-
tis causes trachoma, the leading cause of infectious blindness
Kittens acquire maternal antibodies to C. felis that usually worldwide, and it also is the most common bacterial cause of
protect them from infection until 7 to 9 weeks of age. Both sexually transmitted diseases in humans.22 C. pneumoniae may
humoral and cell-mediated mechanisms provide protective infect nearly all humans, and is an important cause of commu-
immunity in chlamydial infections of other host species. Nat- nity-acquired pneumonia worldwide. It has also been impli-
ural infection seems to confer little protection against reinfec- cated in atherosclerotic cardiovascular disease, although its role
tion, and this protection appears to be short-lived. However, in this condition remains unclear. C. psittaci causes psittacosis
given the low prevalence of chlamydiosis in older cats, an in birds and humans, a potentially life-threatening zoonosis.
age-related resistance to chlamydiosis appears to exist. C. felis has long been suspected as a cause of conjunc-
Both inactivated and attenuated live vaccines are available tivitis in humans. However, many of the reports of human
to protect cats from chlamydial respiratory disease. These infection with C. felis before the advent of genetic detection
vaccines do not entirely prevent infection and shedding of methods remain unconfirmed, because diagnosis was based
chlamydiae after challenge, but they minimize the replica- either on culture, without identification to the species level,
tion of the organism and hence reduce the severity of clini- or on serologic testing, which has the potential to cross-react
cal signs. Transient fever, anorexia, lethargy, and lameness with other chlamydiae and organisms such as Bartonella.
has been described 1 to 3 weeks after use of combined vac- More recently, molecular assays have been used to confirm
cines containing attenuated live C. felis in a low percentage C. felis infection in humans. C. felis was detected in the con-
of cats. Chlamydial vaccines are noncore vaccines that are junctiva of a person with conjunctivitis.24 The organism was
recommended for cats with a high risk of acquiring infec- genetically indistinguishable from an isolate later recovered
tion, such as those introduced to or that reside in catteries from the patient’s cat. Although these associations have been
with documented endemic chlamydiosis. These cats should difficult to document, routine precautions should be taken
be vaccinated regularly after initial treatment of all cats with when handling cats with conjunctivitis, especially young cats
appropriate antimicrobial drugs. with chronic conjunctivitis. Proper hand washing and routine
cleaning and disinfection are likely to be sufficient to prevent
Prevention transmission.
The identification of C. psittaci in dogs with respiratory dis-
Given that chlamydiae survive poorly in the environment and ease and C. pneumoniae in cats with ocular disease suggests
the major route of transmission is close contact, transmission that dogs and cats may have the potential to be a source of
can be minimized by good hygiene, quarantine, single housing, these pathogens for humans, although avian species and other
and disinfection practices in cattery situations. Chlamydiae are infected people, respectively, are likely to be much more signifi-
readily inactivated by detergent solutions. cant sources of infection.

CASE EXAMPLE Eyes, Ears, Nose, and Throat: There was no evidence of ocular
discharge, but mild conjunctival hyperemia was present.
The nasal planum was clean and moist with no discharge;
Signalment: “Sam,” a 3-year-old male neutered domestic no oral erosions or ulcerations were seen.
shorthair cat from Davis, CA Musculoskeletal: Body condition score 7/9. Decreased weight
History: Sam was brought to the University of California, bearing on the left thoracic limb was noted, and the cat
Davis, by a veterinary student for evaluation of fever, sometimes held his left thoracic limb up when sitting. There
right thoracic limb lameness, and bilateral conjunctivitis was possible mild effusion in the left elbow and resentment
with mucopurulent ocular discharge of 2 days’ duration. on flexion of the left carpus and both elbows.
No coughing, sneezing, vomiting, diarrhea, or abnormal All Other Systems: No clinically significant abnormalities were
thirst or urination had been noted. Sam lived indoors with detected.
5 other cats, 3 of which had also had severe conjunctivitis, Laboratory Findings:
ocular discharge, fever, lethargy, and lameness for the CBC:
last week. These 3 cats ranged from 8 months to 2.5 HCT 46.3% (30%-50%)
years of age. All of the cats had been vaccinated regularly MCV 46.9 fL (42-53 fL)
for FHV-1, FCV, and feline panleukopenia virus and MCHC 30.7 g/dL (30-33.5 g/dL)
tested retrovirus-negative as kittens. They were all fed a WBC 10,500 cells/µL (4500-14,000 cells/µL)
commercial dry cat food. Neutrophils 7455 cells/µL (2000-9000 cells/µL)
Physical Examination: Lymphocytes 2310 cells/µL (1000-7000 cells/µL)
Body Weight: 6.7 kg Monocytes 525 cells/µL (50-600 cells/µL)
General: Bright, alert and responsive, hydrated. T = 103.6°F Eosinophils 210 cells/µL (150-1100 cells/µL)
(39.7°C), P = 240 beats/min, respiratory rate 66 breaths/min, Basophils 0 cells/µL (0-50 cells/µL)
mucous membranes pink, CRT = 1 s. Platelets 174,000/µL (180,000-500,000 platelets/µL), with
­moderate macroplatelets.
332 SECTION 2  Bacterial Diseases

Serum Chemistry Profile: Conjunctival Smear Cytology: Chlamydial inclusions were


Sodium 152 mmol/L (151-158 mmol/L) visualized within several epithelial cells.
Potassium 4.4 mmol/L (3.6-4.9 mmol/L) Imaging Findings: Radiographs of the left radius and
Chloride 117 mmol/L (117-126 mmol/L) ulna, including carpus and elbow joints: no radiographic
Bicarbonate 18 mmol/L (15-21 mmol/L) abnormalities.
Phosphorus 4.0 mg/dL (3.2-6.3 mg/dL) Microbiologic Testing: FeLV antigen and FIV antibody
Calcium 9.5 mg/dL (9.0-10.9 mg/dl) serology: negative
BUN 21 mg/dL (18-33 mg/dL) PCR for FCV (oropharyngeal swab specimens): negative
Creatinine 1.6 mg/dL (1.1-2.2 mg/dL) Mycoplasma culture (conjunctival swab specimen): negative
Glucose 151 mg/dL (63-118 mg/dL) Diagnosis: A tentative diagnosis of chlamydial conjunctivitis
Total protein 7.1 g/dL (6.6-8.4 g/dL) and arthritis was made.
Albumin 3.1 g/dL (2.2-4.6 g/dL) Treatment and Outcome: Treatment of all cats in the
Globulin 4.0 g/dL (2.8-5.4 g/dL) household with doxycycline (10 mg/kg PO q24h) was
ALT 48 U/L (27-101 U/L) initiated. This was associated with rapid resolution of fever,
AST 8 U/L (17-58 U/L) ocular signs, and lameness.
ALP 31 U/L (14-71 U/L) Comments: The cats in this household had conjunctivitis and
Gamma GT 0 U/L (0-4 U/L) possibly arthritis in association with chlamydial infection.
Cholesterol 154 mg/dL (89-258 mg/dL) The major differential diagnosis was FCV infection, which
Total bilirubin 0.2 mg/dL (0-0.2 mg/dL). is more widespread than chlamydiosis and for which an
Urinalysis: SGr 1.055; pH 6.5, trace protein, no hemoprotein, association between lameness and infection has been
glucose, ketones or bilirubin, 0-1 WBC/HPF, 0-2 RBC/HPF, few described. Infection with Mycoplasma spp. has also
lipid droplets. been associated with conjunctivitis as well as synovitis
Synovial Fluid Cytology: Synovial fluid from the right stifle, in cats (see Chapter 40). Lameness has developed in cats
right carpus, left carpus, and right tarsus was examined. that were experimentally infected with C. felis, but there
All specimens apart from that from the left carpus were are no reports of lameness in association with natural
markedly blood contaminated and so could not be infection. The negative PCR and culture results for FCV
properly evaluated. The cell counts in those specimens and Mycoplasma spp., respectively, could not definitively
ranged from 2000 to 4000 nucleated cells/µL and consisted rule out infection with these agents. The presence of
predominantly of neutrophils (66%-80%) and small chlamydial inclusions in the conjunctival smear supported
mononuclear cells (7%-18%). The specimen from the left a diagnosis of chlamydiosis, but false positives can occur,
carpus had 2000 nucleated cells/µL, of which 14% were and unfortunately PCR was not performed to confirm
neutrophils, 85% large mononuclear cells, and 1% small the presence of a chlamydial agent. Nevertheless, the
mononuclear cells. There were moderate numbers of rapid response to doxycycline supported the possibility
erythrocytes and blood-associated leukocytes, as well of a bacterial cause of the arthritis. Interestingly,
as low numbers of large mononuclear cells of normal thrombocytopenia was present (and confirmed on smear
morphology, which was consistent with mild blood evaluation), which has been reported in humans with
contamination. chlamydial infections.43

SUGGESTED READINGS 4. Sibitz C, Rudnay EC, Wabnegger L, et  al. Detection of Chla-
mydophila pneumoniae in cats with conjunctivitis. Vet Ophthal-
Gruffydd-Jones T, Addie D, Belak S, et al. Chlamydophila felis infec- mol. 2011;14(suppl 1):67-74.
tion. ABCD guidelines on prevention and management. J Feline Med 5. Sostaric-Zuckermann IC, Borel N, Kaiser C, et al. Chlamydia in canine
Surg. 2009;11:605-609. or feline coronary arteriosclerotic lesions. BMC Res Notes. 2011;4:350.
Laroucau K, Di Francesco A, Vorimore F, et  al. Multilocus variable- 6. Sprague LD, Schubert E, Hotzel H, et  al. The detection of
number tandem-repeat analysis scheme for Chlamydia felis genotyp- Chlamydophila psittaci genotype C infection in dogs. Vet J.
ing: comparison with multilocus sequence typing. J Clin Microbiol. 2009;181:274-279.
2012;50(6):1860-1866. 7. Gresham AC, Dixon CE, Bevan BJ. Domiciliary outbreak of psittacosis
Sibitz C, Rudnay EC, Wabnegger L, et  al. Detection of Chla- in dogs: potential for zoonotic infection. Vet Rec. 1996;138:622-623.
mydophila pneumoniae in cats with conjunctivitis. Vet Ophthalmol. 8. Arizmendi F, Grimes JE, Relford RL. Isolation of Chlamydia psittaci
2011;14(suppl 1):67-74. from pleural effusion in a dog. J Vet Diagn Invest. 1992;4:460-463.
9. Hoelzle K, Wittenbrink MM, Corboz L, et al. Chlamydophila abortus-
REFERENCES induced keratoconjunctivitis in a dog. Vet Rec. 2005;157:632-633.
1. Baker JA. A virus causing pneumonia in cats and producing ele- 10. Sako T, Takahashi T, Takehana K, et al. Chlamydial infection in
mentary bodies. J Exp Med. 1944;79(2):159-172. canine atherosclerotic lesions. Atherosclerosis. 2002;162:253-259.
2. Stephens RS, Myers G, Eppinger M, et  al. Divergence without 11. Lambrechts N, Picard J, Tustin RC. Chlamydia-induced septic
difference: phylogenetics and taxonomy of Chlamydia resolved. polyarthritis in a dog. J S Afr Vet Assoc. 1999;70:40-42.
FEMS Immunol Med Microbiol. 2009;55:115-119. 12. Laroucau K, Di Francesco A, Vorimore F, et  al. Multilocus
3. Dean D, Kandel RP, Adhikari HK, et al. Multiple Chlamydiaceae ­variable-number tandem-repeat analysis scheme for Chlamydia
species in trachoma: implications for disease pathogenesis and con- felis genotyping: comparison with multilocus sequence typing.
trol. PLoS Med. 2008;5:e14. J Clin Microbiol. 2012;50(6):1860-1866.
CHAPTER 33  Chlamydial Infections 333

13. Low HC, Powell CC, Veir JK, et al. Prevalence of feline herpesvirus 30. Dean R, Harley R, Helps C, et  al. Use of quantitative real-time
1, Chlamydophila felis, and Mycoplasma spp. DNA in conjunctival PCR to monitor the response of Chlamydophila felis infection to
cells collected from cats with and without conjunctivitis. Am J Vet doxycycline treatment. J Clin Microbiol. 2005;43:1858-1864.
Res. 2007;68:643-648. 31. Helps C, Reeves N, Tasker S, et  al. Use of real-time quantitative
14. Wills JM, Howard PE, Gruffydd-Jones TJ, et  al. Prevalence of PCR to detect Chlamydophila felis infection. J Clin Microbiol.
Chlamydia psittaci in different cat populations in Britain. J Small 2001;39:2675-2676.
Anim Pract. 1988;29:327-339. 32. Sandmeyer LS, Waldner CL, Bauer BS, et al. Comparison of poly-
15. Kang BT, Park HM. Prevalence of feline herpesvirus 1, feline calici- merase chain reaction tests for diagnosis of feline herpesvirus,
virus and Chlamydophila felis in clinically normal cats at a Korean Chlamydophila felis, and Mycoplasma spp. infection in cats with
animal shelter. J Vet Sci. 2008;9:207-209. ocular disease in Canada. Can Vet J. 2010;51:629-633.
16. Veir JK, Ruch-Gallie R, Spindel ME, et al. Prevalence of selected 33. Borel N, Kempf E, Hotzel H, et  al. Direct identification of chla-
infectious organisms and comparison of two anatomic sampling mydiae from clinical samples using a DNA microarray assay: a vali-
sites in shelter cats with upper respiratory tract disease. J Feline dation study. Mol Cell Probes. 2008;22:55-64.
Med Surg. 2008;10:551-557. 34. Segarra S, Papasouliotis K, Helps C. The in  vitro effects of
17. Maggs DJ, Sykes JE, Clarke HE, et  al. Effects of dietary lysine proxymetacaine, fluorescein, and fusidic acid on real-time
supplementation in cats with enzootic upper respiratory disease. PCR assays used for the diagnosis of feline herpesvirus 1 and
J Feline Med Surg. 2007;9:97-108. ­Chlamydophila felis infections. Vet Ophthalmol. 2011;14
18. Bannasch MJ, Foley JE. Epidemiologic evaluation of multiple (suppl 1):5-8.
respiratory pathogens in cats in animal shelters. J Feline Med Surg. 35. Strom Holst B, Krook L, Englund S, et al. Shedding of chlamydiae
2005;7:109-119. in relation to titers of serum chlamydiae-specific antibodies and
19. Helps CR, Lait P, Damhuis A, et  al. Factors associated with serum concentrations of two acute-phase proteins in cats without
upper respiratory tract disease caused by feline herpesvirus, conjunctivitis. Am J Vet Res. 2011;72:806-812.
feline calicivirus, Chlamydophila felis and Bordetella bronchi- 36. Ohya K, Okuda H, Maeda S, et al. Using CF0218-ELISA to distin-
septica in cats: experience from 218 European catteries. Vet Rec. guish Chlamydophila felis-infected cats from vaccinated and unin-
2005;156:669-673. fected domestic cats. Vet Microbiol. 2010;146:366-370.
20. von Bomhard W, Polkinghorne A, Lu ZH, et  al. Detection of 37. Sparkes AH, Caney SM, Sturgess CP, et  al. The clinical efficacy
novel chlamydiae in cats with ocular disease. Am J Vet Res. of topical and systemic therapy for the treatment of feline ocular
2003;64:1421-1428. chlamydiosis. J Feline Med Surg. 1999;1:31-35.
21. Richter M, Matheis F, Gonczi E, et al. Parachlamydia acantham- 38. Donati M, Piva S, Di Francesco A, et al. Feline ocular chlamydiosis:
oebae in domestic cats with and without corneal disease. Vet Oph- clinical and microbiological effects of topical and systemic therapy.
thalmol. 2010;13:235-237. New Microbiol. 2005;28:369-372.
22. Saka HA, Thompson JW, Chen YS, et al. Quantitative proteomics 39. Sturgess CP, Gruffydd-Jones TJ, Harbour DA, et  al. Controlled
reveals metabolic and pathogenic properties of Chlamydia tracho- study of the efficacy of clavulanic acid-potentiated amoxicil-
matis developmental forms. Mol Microbiol. 2011;82:1185-1203. lin in the treatment of Chlamydia psittaci in cats. Vet Rec.
23. Gruffydd-Jones T, Addie D, Belak S, et  al. Chlamydophila felis 2001;149:73-76.
infection. ABCD guidelines on prevention and management. 40. Gerhardt N, Schulz BS, Werckenthin C, et  al. Pharmacokinetics
J Feline Med Surg. 2009;11:605-609. of enrofloxacin and its efficacy in comparison with doxycycline in the
24. TerWee J, Sabara M, Kokjohn K, et  al. Characterization of the treatment of Chlamydophila felis infection in cats with conjunctivitis.
systemic disease and ocular signs induced by experimental infection Vet Rec. 2006;159:591-594.
with Chlamydia psittaci in cats. Vet Microbiol. 1998;59:259-281. 41. Hartmann AD, Helps CR, Lappin MR, et  al. Efficacy of prado-
25. Dickie CW, Sniff ES. Chlamydia infection associated with peritoni- floxacin in cats with feline upper respiratory tract disease due to
tis in a cat. J Am Vet Med Assoc. 1980;176:1256-1259. Chlamydophila felis or Mycoplasma infections. J Vet Intern Med.
26. Hargis AM, Prieur DJ, Gaillard ET. Chlamydial infection of the 2008;22:44-52.
gastric mucosa in twelve cats. Vet Pathol. 1983;20:170-178. 42. Owen WM, Sturgess CP, Harbour DA, et al. Efficacy of azithro-
27. Wills JM. Chlamydial infection in the cat. University of Bristol: mycin for the treatment of feline chlamydophilosis. J Feline Med Surg.
PhD Thesis; 1986. 2003;5:305-311.
28. O’Dair HA, Hopper CD, Gruffydd-Jones TJ, et al. Clinical aspects 43. Day CJE, Fawcett IW. Psittacosis and acute thrombocytopenic pur-
of Chlamydia psittaci infection in cats infected with feline immuno- pura. J R Soc Med. 1992;85(6):360-361.
deficiency virus. Vet Rec. 1994;134:365-368.
29. Sykes JE, Studdert VP, Browning GF. Comparison of the poly-
merase chain reaction and culture for the detection of feline Chla-
mydia psittaci in untreated and doxycycline-treated experimentally
infected cats. J Vet Intern Med. 1999;13:146-152.
CHAPTER 34

Streptococcal and Enterococcal Infections


Jane E. Sykes

humans, streptococci that infect dogs and cats range from com-
Overview of Streptococcal and Enterococcal mensals of low virulence through to highly virulent organisms
Infections that can cause severe disease manifestations and death.
First Described: Streptococci were first described in Germany Streptococci have been classified based on their hemolytic
in 1868 by Billroth.1 Enterococci were separated from the properties into β-hemolytic streptococci, which cause com-
genus Streptococcus in 1984.2 plete hemolysis when they grow on blood agar; γ-hemolytic or
nonhemolytic streptococci, which do not cause hemolysis; and
Major Causes: In dogs and cats, the most common causes
α-hemolytic streptococci, which reduce hemoglobin, causing a
are Streptococcus canis, Streptococcus equi subsp. zooepi-
greenish discoloration of the agar around bacterial colonies.
demicus, Enterococcus faecium, and Enterococcus faecalis,
Streptococci are also classified from A through W based on their
which are gram-positive coccoid bacteria
surface antigens (Lancefield classification system) and classi-
Geographic Distribution: Worldwide fied based on phenotypic characteristics (such as pyogenic or
Mode of Transmission: Direct contact. Organisms are usually viridans group streptococci). Major pathogenic streptococci of
commensals that invade opportunistically. dogs and cats belong to Lancefield groups B, C, D, or G (Table
34-1). Pyogenic streptococci are β-hemolytic streptococci that
Major Clinical Signs: Neonatal septicemia, pharyngitis, cervi- belong to Lancefield groups A (S. pyogenes), B (Streptococcus
cal lymphadenitis, bacteremia and endocarditis, urinary agalactiae), C (which includes Streptococcus dysgalactiae and
tract infections, postoperative incision or wound infec- S. equi subsp. zooepidemicus), and G (which includes Strepto-
tions, otitis externa, keratitis, bronchopneumonia, pyome- coccus canis). Viridans streptococci are often nonhemolytic or
tra or metritis, meningoencephalitis, necrotizing fasciitis α-hemolytic (viridis = Latin for green) and tend to be commen-
and toxic shock syndrome (primarily S. canis), rhinitis and sals that have low virulence and rarely invade tissues opportu-
necrotizing sinusitis, pyothorax, discospondylitis, arthritis, nistically. Many group D streptococci have now been classified
osteomyelitis, mastitis, cholangiohepatitis, and peritonitis as enterococci. Molecular typing schemes have revealed that
Differential Diagnoses: Primarily other gram-positive and gram- hemolytic reactions, Lancefield antigens, and phenotypic char-
negative bacterial infections acteristics do not necessarily predict genetic relatedness among
Human Health Significance: Streptococcal species that infect
dogs and cats occasionally infect human patients. Dogs
and cats may also have the potential to serve as sources of
multidrug-resistant enterococcal infections for humans.

Etiology, Epidemiology, and Clinical Features


Streptococci are catalase-negative, gram-positive cocci that
divide along a single axis, forming pairs and chains of organ-
isms (Figures 34-1 and 34-2). They are facultative to strict
anaerobes that require complex media for growth, preferably
supplemented with blood. More than 40 species of streptococci
have been described. Species of streptococci vary in host tro-
pism and virulence properties, and so it is important that they
be discussed separately. One of the most important streptococ-
cal pathogens of humans, Streptococcus pyogenes, which causes
streptococcal pharyngitis (“strep throat”), does not infect dogs FIGURE 34-1  Histopathology of the serosal surface of the liver of a 12-day-old
and cats to a significant degree (see Public Health Aspects). domestic shorthair kitten that was euthanized after it was found hypothermic and
However, interspecies transmission has been reported for some tachypneic. Severe suppurative omphalophlebitis and peritonitis was present. Lesions
pathogenic streptococci such as Streptococcus pneumoniae contained numerous chains of gram-positive cocci. Brown and Benn stain, 1000× oil
and Streptococcus equi subsp. zooepidemicus. As is the case in magnification.

334
CHAPTER 34  Streptococcal and Enterococcal Infections 335

streptococcal strains. Nevertheless, these traditional classifica- Streptococcus canis


tion methods are still used by laboratory staff, microbiologists, S. canis is the most frequently isolated streptococcus from dogs
and clinicians to describe streptococci. and cats.3 It is a β-hemolytic, group G (pyogenic) streptococ-
Streptococci invade tissues opportunistically when there is a cus that colonizes the skin, genital, and gastrointestinal tracts
breach in normal host barriers. This leads to a variety of clinical of healthy dogs and cats. It has also been isolated from other
disease manifestations, such as pyoderma, pneumonia, endo- animal species, such as rats, mice, rabbits, mink, and foxes.
carditis, arthritis, osteomyelitis, meningoencephalitis, cellulitis, Infection with S. canis may be associated with neonatal bacte-
and urinary tract infections (UTIs). Severe and life-threatening remia, pharyngitis, cervical lymphadenitis, infective endocar-
manifestations of streptococcal infection include necrotizing ditis, UTIs, postoperative incision or wound infections, otitis
fasciitis (NF) and streptococcal toxic shock syndrome (STSS). externa, keratitis, bronchopneumonia, pyometra or metritis,
Predisposing conditions for streptococcal infections in dogs meningoencephalitis, NF, STSS, rhinitis and necrotizing sinus-
and cats may include atopic dermatitis, wounds, foreign bodies itis, pyothorax, discospondylitis, arthritis, osteomyelitis, masti-
(such as migrating grass awns), immunosuppressive drug treat- tis, cholangiohepatitis, and peritonitis. Neonatal infections can
ment, and young age (especially neonates).3 The most common occur when organisms are transmitted from the vaginal tract
clinical presentation among fetuses and neonates is streptococ- during parturition. The organism can then gain access to the
cal bacteremia and sepsis, which can result in abortion and systemic circulation via the umbilical vein. Streptococcal men-
death. Streptococcal pneumonia may occur in association with ingitis results from direct extension from the sinuses or middle
infections with other respiratory pathogens, such as respiratory ear, or from bacteremia. Other embolic complications may
viruses, or occur secondary to bronchitis, hematogenous spread accompany bacteremia with group G streptococci. Although
or aspiration. In one study, streptococci were isolated from sick opportunistic infections with S. canis occur sporadically, out-
dogs more frequently in summer months.3 breaks of group G streptococcal infection have been reported
in group-housed animals, which suggested spread of a virulent
strain.4-6
Severe manifestations of S. canis infection, such as STSS and
NF, have been increasingly described in dogs and cats in recent
years, sometimes in the absence of obvious immunosuppressive
underlying conditions or wounds.6-9 Although toxic shock syn-
drome can also be caused by staphylococci, STSS is defined as
any streptococcal infection associated with the sudden onset of
shock and organ failure. Mechanisms of shock and organ fail-
ure identified in human group A streptococcal infections include
elaboration of pyrogenic exotoxins by streptococci, which act as
A B superantigens. Superantigens stimulate T cell responses through
their ability to bind to, and cross-link, the MHC class II com-
FIGURE 34-2  Electron micrograph of Streptococcus pyogenes (A) and Streptococcus
canis (B). Proteinaceous cell surface fibrillae are present on S. canis that resemble the plex of antigen-presenting cells, and the T cell receptor, which
M-protein surface fibrils of the human pathogen S. pyogenes. M-protein is a key virulence bypasses normal MHC-restricted antigen processing. This leads
factor of S. pyogenes. (From DeWinter LM, Low DE, Prescott JF. Virulence of Streptococcus to sudden and massive cascade of cytokine release, which in
canis from canine streptococcal toxic shock syndrome and necrotizing fasciitis. Vet Micro- turn causes signs of fever, vomiting, and hypotension, together
biol 1999;70:95-110.) with tissue damage, disseminated intravascular coagulation

TABLE 34-1
Classification of Streptococci Isolated from Dogs and Cats
Lancefield Hemolytic
Species Antigen(s) Reactions Comments
S. canis G β Most common. Dogs and cats are natural hosts. Causes a variety
of clinical syndromes. Primary cause of streptococcal toxic shock
syndrome and necrotizing fasciitis in dogs and cats.
S. equi subsp. C β Causes pneumonia and sinusitis, primarily in group-housed animals.
zooepidemicus Has also been associated with meningitis.
S. dysgalactiae C β Uncommon. Has been isolated from dogs with septicemia, dermatitis,
or pneumonia.
S. bovis group D α or γ Uncommon. Has been isolated from dogs with urinary tract infections
(S. gallolyticus and and endocarditis.
S. infantarius)
S. agalactiae B β or γ Rare. Isolated from dogs and cats with endocarditis and neonatal
septicemia.
336 SECTION 2  Bacterial Diseases

(DIC), and multiple organ dysfunction.10 Other streptococcal “flesh-eating bacteria” has been used to describe the organisms
virulence factors also contribute to proinflammatory cytokine involved. Streptococcal NF has been described in both dogs and
release and the development of hypotension. Laboratory abnor- cats. Lesions usually involve a limb and are intensely painful,
malities include thrombocytopenia, azotemia, hypoalbumin- with localized heat and swelling and accumulation of exudate
emia, and metabolic acidosis. Death can occur within 48 hours along fascial planes that requires drainage and debridement. In
after the onset of illness. Specific criteria are used for diagno- some dogs, extensive sloughing of necrotic skin occurs.9 Nec-
sis of STSS in human patients, and similar definitions could be rotizing myositis, bacteremia, and septic emboli may accom-
used for diagnosis of STSS in dogs and cats (Box 34-1). Because pany NF.8 Outbreaks of NF, arthritis, sinusitis, and meningitis
only a few dogs and cats with STSS have been described, pre- caused by S. canis have been reported in cats in shelters5 and
disposing factors have not been clearly identified. Predisposing breeding colonies.11 Molecular typing (by pulsed-field gel elec-
conditions in humans include diabetes mellitus, alcoholism, sur- trophoresis) of isolates from one outbreak revealed that iso-
gical procedures, penetrating and nonpenetrating trauma, viral lates were clonally related, which indicated spread of a virulent
infections such as varicella, and possibly the use of nonsteroidal strain.12
antiinflammatory drugs. Despite the recognition of severe disease manifestations in
NF is a bacterial infection of the deep subcutaneous tissues some dogs and cats, relatively little is known about virulence
and fascia, characterized by extensive necrosis and gangrene factors of S. canis. A protein analogous to M protein, a major
of the skin and underlying tissues. NF often begins as a minor virulence factor of S. pyogenes, has been identified in S. canis
wound and progresses rapidly over 24 to 72 hours, and it may and was shown to bind plasminogen and degrade thrombi.13
be accompanied by STSS (Figure 34-3). The popular term M protein has important antiphagocytic properties. Genes that
encode M protein and a streptococcal hemolysin, streptolysin
O, have been detected in S. canis isolates from dogs with NF
and STSS.14 Genes that encode other virulence factors identi-
BOX 34-1 fied in S. pyogenes, such as pyrogenic exotoxins (Spe genes),

Criteria Used for Diagnosis of STSS in Human Patients,


with Suggested Modifications for Diagnosis of STSS in Dogs
and Cats*

I  Isolation of Group G streptococci


(Streptococcus canis)
A. From a normally sterile site
B. From a nonsterile site

II  Clinical signs of severity


A. Hypotension: systolic blood pressure <90 mm Hg in
dogs and <80 mm Hg in cats
and
B. Two or more of the following signs:
1. Renal impairment: creatinine above the upper limit
of the reference range A
2. Coagulopathy: thrombocytopenia or DIC (prolonged
clotting times, low serum fibrinogen concentration,
and increased fibrin degradation products)
3. Liver involvement: serum AST, ALT, or total biliru-
bin levels at least twice the upper limit of the refer-
ence range
4. Acute respiratory distress syndrome defined as acute
onset of diffuse pulmonary infiltrates and hypox-
emia in the absence of cardiac failure, or evidence
of diffuse capillary leak manifested as acute onset of
pulmonary edema, or pleural or peritoneal effusions
with hypoalbuminemia
5. A generalized erythematous macular rash that may
desquamate
6. Soft tissue necrosis, including necrotizing fasciitis or
myositis, or gangrene B
FIGURE 34-3  A, Two-year-old female spayed domestic shorthair that was eutha-
*An illness that fulfills criteria IA and II is defined as a definite case. nized with necrotizing cellulitis, myositis and fasciitis, and peritonitis that developed after
An illness that fulfills criteria IB and II is defined as a probable case if an ovariohysterectomy site became infected with Streptococcus canis. B, Histopathology
no other cause for the illness is identified. showed severe necrotizing cellulitis, myositis, and cellulitis with intralesional cocci that
DIC, disseminated intravascular coagulation; STSS, streptococcal toxic sometimes formed chains. H&E stain, 1000× oil magnification. (Courtesy of the University
shock syndrome. of California, Davis, Veterinary Anatomic Pathology Service.)
CHAPTER 34  Streptococcal and Enterococcal Infections 337

streptococcal superantigen (SSA), streptokinase (Ska), and C5a zooepidemicus meningoencephalitis occurred in a cat as a result
peptidase (Scp, which cleaves the fifth component of comple- of extension of otitis media/interna.21
ment) have not been detected. The pathogenesis of infection with S. equi subsp. zooepi-
demicus in dogs and cats is unclear. Co-infection with other
Streptococcus equi subsp. zooepidemicus contagious respiratory pathogens can contribute to the sever-
S. equi subsp. zooepidemicus is a β-hemolytic, group C strepto- ity of disease. Co-infections with canine distemper virus
coccus that is a commensal of the upper respiratory and lower (CDV), canine adenovirus-2 (CAV-2), canine herpesvirus-1
genital tracts of horses, and appears to be an emerging pathogen (CHV-1), canine influenza virus (CIV), Bordetella bronchi-
of dogs and cats. Opportunistic infections such as wound infec- septica, or Mycoplasma cynos have been documented in
tions, respiratory disease, endometritis, and abortion occur in some, but not all, affected dogs with S. equi subsp. zooepi-
horses. Infections have also been reported in a variety of other demicus pneumonia. Experimentally, co-infection of dogs
animal species, including cattle, sheep, pigs, monkeys, seals, with both CIV and S. equi subsp. zooepidemicus is asso-
ruminants, dogs, cats, and humans. Different strains of S. equi ciated with more severe clinical manifestations of disease
subsp. zooepidemicus exist that may vary in pathogenicity and than when either organism is inoculated alone.22 Other
host tropism. Infected horses may have been a source of infec- environmental factors such as overcrowding may also con-
tion for dogs in some situations,15,16 but in other situations tribute to stress and severe disease manifestations. Supe-
where infected dogs and cats have been identified, a history of rantigen genes have been detected in some isolates of
horse contact was not present. S. equi subsp. zooepidemicus from dogs,23,24 but there has
Several outbreaks of hemorrhagic, fibrinosuppurative, and been no correlation between the presence of these genes and
necrotizing pneumonia associated with S. equi subsp. zooepi- the severity of histopathologic lesions. However, expression
demicus infection have been described in group-housed dogs, of proinflammatory cytokines such as TNF-α, Il-6, and Il-8 in
such as those in kennels and shelter environments, from a vari- the lungs of affected dogs suggests that a potent inflammatory
ety of geographic locations worldwide (Figure 34-4).17 The response is involved in the pathogenesis of disease.23
pneumonia progresses rapidly in some dogs can be accompa-
nied by signs suggestive of STSS, such as fever and systemic Other Streptococcal Species
hypovolemia. In some dogs, severe tachypnea and death occurs Like S. equi subsp. zooepidemicus, S. dysgalactiae is a β-hemolytic,
within 48 hours of the first signs of respiratory disease, and dogs group C streptococcus. There are two subspecies, S. dysgalactiae
may be found dead without any preceding clinical signs. Other subsp. equisimilis and S. dysgalactiae subsp. dysgalactiae. After
infected dogs show only mild signs of respiratory disease. S. equi S. canis and S. equi subsp. zooepidemicus, S. dysgalactiae subsp.
subsp. zooepidemicus bacteremia and sepsis has been reported equisimilis was the third most common β- or γ-hemolytic strep-
in racing greyhounds.18 Chronic lymphoplasmacytic rhinitis, tococcal species isolated from dogs at a veterinary teaching hos-
sometimes with turbinate lysis, has also been described in sev- pital in the United States, and was detected in 13 (3.3%) of 393
eral dogs in association with S. equi subsp. zooepidemicus infec- dogs with streptococcal infections. Affected dogs had bacteremia
tion, which resolved after specific antimicrobial treatment.15,16 and sepsis, dermatitis, or pneumonia.3 S. dysgalactiae subsp. dys-
Outbreaks of pneumonia, purulent rhinosinusitis, and menin- galactiae has been reported in association with systemic neonatal
goencephalitis have been described in cats.19,20 S. equi subsp. infections and death in a litter of great Dane puppies.25
The porcine streptococcus, Streptococcus suis, has been
isolated from cats with meningoencephalitis, pyothorax, or
dermatitis.26,27 S. suis was also isolated from the urine of a sys-
temically unwell dog in Canada28 and can be isolated from the
tonsils, anus, and skin of healthy dogs and cats.29 Although con-
tact with pigs was not reported in the sick dogs and cats, the dog
was fed commercial pig ear treats. S. suis has been classified into
serotypes, which have epidemiologic importance. Human infec-
tions are usually serotypes 2 and 14. The cases reported in dogs
and cats have not belonged to these serotypes.
Other streptococcal species isolated from dogs and cats
include the human pathogen Streptococcus constellatus (a Strep-
tococcus anginosus group organism), which was isolated from a
dog with pyoderma;30 and group B streptococci (S. agalactiae),
which have been isolated from dogs and cats with streptococcal
disease such as endocarditis and neonatal sepsis. Group B strepto-
cocci are important causes of puerperal sepsis and neonatal sepsis
and meningitis in humans, as well as mastitis in cattle.

Streptococcus bovis Group (Streptococcus


gallolyticus and Streptococcus infantarius)
Streptococcus bovis group organisms are α- or nonhemolytic and
usually possess Lancefield group D antigens. Biotypes that belong
FIGURE 34-4  Pneumonia in a shelter dog caused by Streptococcus equi subsp. zooepi- to this group, which differ in their biochemical characteristics,
demicus. The lungs are consolidated and dark red. (Courtesy of Dr. Patricia Pesavento, Uni- have now been reclassified as distinct species, Streptococcus gallo-
versity of California, Davis.) lyticus and Streptococcus infantarius.31 S. bovis group organisms
338 SECTION 2  Bacterial Diseases

are commensals of the gastrointestinal tracts of humans and and virulence properties. E. faecium is more likely to show high-
other animals, especially ruminants. They are important causes level resistance to penicillins and carbapenems than E. faecalis,
of bacteremia and endocarditis in humans, accounting for 11% and as a result, in humans, the prognosis for serious E. faecium
to 17% of all endocarditis cases. They frequently affect mul- infections may be worse than that for E. faecalis infections.40
tiple valves and are associated with embolic complications. For However, E. faecalis is more likely to produce biofilms than
unknown reasons, fecal carriage of S. bovis group organisms by E. faecium.41 Although most infections in humans have been
humans has been strongly associated with colorectal cancer. This E. faecalis, a worrisome increase in the prevalence of MDR E.
especially seems to be true for S. gallolyticus subsp. gallolyticus.32 faecium infections has occurred. In particular, ampicillin- and
Rarely, S. bovis group organisms are isolated from dogs and cats fluoroquinolone-resistant E. faecium strains that belong to hos-
with UTIs. They have also been isolated from the blood of dogs pital adapted strains known as clonal complex 17 (CC17) have
with mitral valve endocarditis, one of which also had sternebral emerged. CC17 isolates have been found in dogs, but their viru-
osteomyelitis.33,34 lence gene profile and antimicrobial susceptibility profiles have
differed from those of human isolates.42
Enterococcus spp. In healthy dogs and cats, enterococci can be found on the
Enterococci are α- or nonhemolytic gram-positive bacteria that skin and within the oral cavity, nasal cavity, and gastrointesti-
usually possess Lancefield group D antigens, are commensals of nal tract. In the United States, the most prevalent species in dogs
the gastrointestinal tracts of humans and other animals, and are appears to be E. faecalis, which comprised 68% of 155 iso-
important nosocomial pathogens. They form pairs or short chains lates.43 In the cat, it was Enterococcus hirae, which comprised
of organisms and cannot morphologically be discriminated from 52% of 121 isolates.43 Preparations that contain E. faecium
streptococci. Enterococci survive harsh environmental condi- strain SF68 have been investigated as probiotics in dogs and
tions; they grow in medium that contains 6.5% NaCl, can grow cats, with mixed results.44-46 Some studies suggest that supple-
at temperatures that range from 10°C to 45°C (50°F to 113°F), mentation of food with this probiotic can stimulate local and
and hydrolyze esculin in the presence of 40% bile salts. The genus systemic immune function in dogs and cats.47,48
Enterococcus was separated from Streptococcus in the mid-1980s Although less pathogenic than many streptococci, impor-
based on the results of DNA hybridization experiments.35 tant virulence factors have been identified in enterococci that
Enterococci are often resistant to a wide variety of different enable them to invade tissues and cause disease. These include
antimicrobial drug classes, and multiple drug resistant (MDR) a cytolysin, gelatinase, and a serine protease, as well as several
enterococcal infections, especially those that are also resistant surface proteins (e.g., the Ace protein, which allows the organ-
to vancomycin (vancomycin-resistant enterococci or VRE), ism to adhere to connective tissue components, such as colla-
are a significant problem in human medicine. Of great clinical gen). Enterococci also possess pili that play an important role
importance, resistance among enterococci occurs as a result of in adherence. The ability of many enterococci to form biofilms
both intrinsic and acquired resistance mechanisms. Intrinsic means they can be very difficult to eradicate, because they resist
resistance to low levels of most β-lactam antimicrobials occurs phagocytosis and antimicrobial drugs.41 As in humans, the two
because enterococci possess low-affinity penicillin binding pro- most common species of Enterococcus associated with disease
teins (PBPs). Thus penicillins are generally bacteriostatic, not in dogs and cats are E. faecalis and E. faecium.36,49 Others are
bactericidal, against enterococci. Enterococci also have low- shown in Box 34-2.
level intrinsic resistance to aminoglycosides, which results from
decreased drug uptake. However, uptake of aminoglycosides
is enhanced when enterococci are exposed to β-lactams, which
explains the synergistic activity of this combination. Enterococci
are resistant to trimethoprim-sulfamethoxazole because they
BOX 34-2
utilize exogenously produced folate. Thus, even if the organ- Enterococcus Species Isolated from Healthy and Sick
isms appear sensitive to trimethoprim-sulfamethoxazole in vitro
Dogs and Cats36,43,50,51
(when exogenous folate is lacking), the drug is not effective
in  vivo. Many laboratories therefore do not report minimum
inhibitory concentrations for this organism-drug combination. Healthy Dogs and Cats
Enterococcal resistance to other antimicrobials, such as mac- E. faecalis
rolides and vancomycin, can result from acquired resistance E. faecium
mechanisms. Extensive multidrug resistance has been well doc- E. hirae
umented in enterococci isolated from dogs and cats, but there E. casseliflavus
are only a few reports of enterococci with acquired vancomycin E. durans
resistance from dogs in the literature. These have been from the E. canintestini
United Kingdom, New Zealand, and the United States.36-38 All E. avium
other isolates examined have been vancomycin susceptible. In
contrast, VRE account for about 30% of enterococcal infec- Sick Dogs and Cats
tions in humans from Europe and North America.39 E. faecalis
Clinical microbiology laboratories often do not differentiate E. faecium
between Enterococcus species, because definitive identification E. hirae
can require genetic typing. Some laboratories use biochemical E. casseliflavus
methods to identify Enterococcus faecalis and Enterococcus E. durans
faecium, which are the most prevalent species in humans and E. gallinarum
which differ in their epidemiology, antimicrobial susceptibility,
CHAPTER 34  Streptococcal and Enterococcal Infections 339

Because enterococci are naturally resistant to a number accompanied by thrombocytopenia as a result of DIC, azote-
of antimicrobials, treatment with broad-spectrum antibiotics mia and/or increased liver enzyme activities, hypoalbuminemia,
(which destroy competing bacteria within the gastrointestinal and hypoglycemia. The urinalysis of dogs and cats with strep-
tract) selects for colonization by enterococci. The development tococcal or enterococcal UTIs may reveal pyuria, proteinuria,
of enterococcal UTIs may be promoted by repeated use of anti- or hematuria, and cocci in pairs or chains may sometimes be
microbial drugs to treat recurrent UTIs, when the underlying evident on sediment examination.
cause of infection is not or cannot be resolved. Enterococci can
contaminate the hospital environment and survive for long peri- Clotting Function
ods on fomites such as doorknobs, stethoscopes, and monitoring Dogs and cats with STSS that develop DIC may have clotting
devices, as well as the hands, gloves, and clothing of hospital per- function abnormalities including prolonged coagulation times,
sonnel. Inoculation may occur via urinary, intravenous, or other increased fibrin degradation product and D-dimer concentra-
invasive devices, or organisms may enter the body from a dam- tions, and decreased antithrombin concentrations.
aged gastrointestinal tract. Severe concurrent illness and immuno-
suppression may predispose to infection. In addition to skin and Cytologic Abnormalities
UTIs, enterococci may be isolated from dogs and cats with UTIs, Cytologic examination of specimens (respiratory lavage fluid,
pyoderma and otitis externa, cholangiohepatitis, pancreatitis, aspirates of affected tissues, or pus) from dogs and cats with
hepatic abscesses, peritonitis, mastitis, bacteremia and endocardi- streptococcal or enterococcal infections typically reveals large
tis, wound infections, and discospondylitis. There have been rare numbers of degenerate and nondegenerate neutrophils. Coccoid
reports of gastrointestinal illness in association with enterococcal bacteria in pairs or chains may also be seen.
infection of the gastrointestinal tract in dogs and cats. In affected
cats, an association with diarrhea was supported by the presence Microbiologic Tests
of large mats of enterococci in association with the gastrointesti- Isolation and Identification
nal mucosa.52,53 Frequently, enterococci are isolated from lesions Streptococci and enterococci that infect dogs and cats are read-
in mixed infections with other gastrointestinal microorganisms. ily isolated on routine bacteriologic media from aspirates, tis-
sue biopsies, body fluids, and lavage specimens. Inoculation of
Physical Examination Findings complex media that contain blood is recommended. Streptococci
Physical examination findings in dogs and cats with Streptococ- grow well in anaerobic incubation conditions, but these are not
cus or Enterococcus infections vary with the anatomic site(s) of required because of their facultative nature. The laboratory may
infection. Streptococcal infections are an important differential use selective media for specimens that are heavily contaminated
diagnosis in sick or fading neonates; dogs that have physical with other bacteria. Species are identified based on their pheno-
examination findings suggestive of endocarditis; dogs and cats typic characteristics (such as colony morphology and hemolytic
with severe pneumonia and pyothorax; NF; rapid onset of sys- properties), biochemical reactions, and serotyping with Lance-
temic illness in association with a wound; or outbreaks of severe field antisera; the last is available on a commercial basis. As
purulent upper respiratory disease, hemorrhagic pneumonia, noted previously, genetic typing may be required to identify some
and systemic illness in group-housed animals. enterococci to the species level. Molecular typing methods such
Findings suggestive of streptococcal or enterococcal endocar- as pulsed-field gel electrophoresis may be required to investigate
ditis include fever, lethargy, tachycardia, joint pain and swelling, outbreaks in group-housed animals or hospital environments.
systolic heart murmurs, and signs of embolic complications such as Antimicrobial susceptibility testing for streptococci is often
lameness, limb edema, weak or absent pulses, or neurologic signs. not performed by veterinary diagnostic laboratories, because vir-
Animals with streptococcal pneumonia may have a fever (up to tually all strains are penicillin susceptible. Group A streptococci
106.3°F or 41.3°C), cough, tonsillar enlargement and hyperemia, (GAS) that infect humans remain universally susceptible to peni-
mucopurulent nasal discharge, and/or tachypnea with increased cillin, but an increased prevalence of penicillin resistance, which
lung sounds. Lung sounds may be decreased if pleural effusion is occurs as a result of altered PBPs, has been detected in S. pneu-
present. Animals with NF have variable extents of regional cutane- moniae, which is primarily a human pathogen.54 S. pneumoniae
ous erythema, sometimes in association with a wound; cutaneous may also be resistant to other β-lactam drugs and macrolides.
ulceration, swelling, edema, intense pain, and warmth on palpa- Evaluation of antimicrobial susceptibility is of critical importance
tion of the affected area. In some animals, extensive skin sloughing for enterococci, because of the high prevalence of resistance in
occurs. Animals with STSS may be obtunded, with weak pulses, this group, and especially among E. faecium isolates. Susceptibil-
tachycardia, tachypnea, and prolonged capillary refill time. Rectal ity testing for aminoglycosides requires high-concentration disk
temperatures may exceed 106°F (41.1°C), although the presence diffusion or broth microdilution methods, which may not rou-
of fever is variable. Icterus may be present in animals that develop tinely be performed by some veterinary microbiology laboratories
multiple organ dysfunction syndrome in association with STSS. (see Treatment).

Diagnosis Pathologic Findings


Pathologic findings in Streptococcus infections vary depending
Laboratory Abnormalities on the organ system affected. Gross findings in S. canis sinusitis
Complete Blood Count, Biochemistry, and Urinalysis Findings include accumulation of purulent material within the frontal
Dogs and cats with severe manifestations of streptococcal infec- sinus with osteolysis, and distortion of the nasal bridge in cats.5
tions, such as pneumonia, NF, endocarditis, and STSS, usually When meningitis is present, purulent material may be found
have a neutrophilic leukocytosis with a left shift, toxic neutro- in association with the meninges (Figure 34-5). Lesions of NF
phils, lymphopenia, and monocytosis. A degenerative left shift in dogs and cats consist of edema, ecchymoses, and necrosis
or circulating metamyelocytes may be present. STSS may be that extend within the fascia and subcutis in association with
340 SECTION 2  Bacterial Diseases

FIGURE 34-5  Accumulation of purulent exudate over the meninges in a shelter cat
that died of Streptococcus canis meningitis. (Courtesy of Dr. Patricia Pesavento, University FIGURE 34-6  Histopathology of the mitral valve of an 8-year-old male neutered
of California, Davis.) springer spaniel that was seen for a 1-month history of right thoracic limb lameness,
anorexia, and lethargy. There is severe, subacute, fibrinonecrotic endocarditis with intra-
ulcerative skin lesions (see Figure 34-3). An accumulation of lesional cocci. When the dog was first evaluated, the platelet count was 9000 platelets/
µL and the dog was treated with vincristine for immune-mediated thrombocytopenia.
pus may be found between fascial planes. Congestion of a
Subsequently the dog developed vomiting and diarrhea and became obtunded. Blood
variety of organs may be present in animals that have accom- cultures grew Streptococcus canis. At necropsy, septic emboli were present in the kidneys,
panying STSS. When S. canis or Enterococcus endocarditis is spleen, and lymph nodes. Necrotizing fasciitis was also present in multiple limbs, which
present, adherent and friable mass lesions may be seen in asso- was thought to have developed as a result of septic embolization.
ciation with the mitral and/or, less commonly, the aortic valves.
Dogs with streptococcal endocarditis are more likely to have
concurrent polyarthritis than dogs with other bacterial causes may also have synergistic activity against some streptococci,
of endocarditis.33 On histopathology, valve lesions generally such as group C streptococci (S. equi subsp. zooepidemicus).
contain large numbers of degenerate and nondegenerate neu-
trophils, hemorrhage, necrosis, fibrin thrombi, and abundant Necrotizing Fasciitis, Myositis,
gram-positive cocci (Figure 34-6). Septic thromboemboli may and Toxic Shock Syndrome
be present in a variety of organs. Histopathologic lesions in Prompt surgical exploration and debridement is of critical
neonates that die of neonatal sepsis include severe inflamma- importance in NF, because shock and organ failure will con-
tion and necrosis, as well as septic emboli in a variety of tissues tinue to progress in the face of antimicrobial treatment when
(see Figure 34-1). necrotic tissue persists. Imaging techniques such as ultrasound
Grossly, the lungs of dogs with S. equi subsp. zooepidemicus may be useful to help characterize the extent of lesions before
pneumonia may be red, consolidated, and fail to collapse (see surgery. Extremely aggressive fluid resuscitation with crystal-
Figure 34-4). There may be variable quantities of hemorrhagic loids and blood products may also be required, while blood
pleural effusion, petechial and ecchymotic hemorrhages on pleu- pressure is monitored. In human patients, deep-seated strep-
ral surfaces, and bloody exudate within the airways and nasal tococcal infections (NF and STSS) are treated with both high-
cavity.17 Typical histopathologic changes include fibrinous, nec- dose penicillin and clindamycin. This is because clindamycin
rotizing, and/or suppurative pneumonia or bronchopneumonia; suppresses exotoxin production by GAS, is more active than
fibrin thrombi; and intralesional clusters or chains of coccoid penicillin in experimental NF, and has a longer half-life than
bacteria. Within lymph nodes, marked lymphoid depletion may penicillin. However, clindamycin resistance exists in some GAS,
be present, but lymphoid hyperplasia has also been reported. whereas all GAS are susceptible to penicillin.57 Also, penicil-
Occasionally, septic thromboemboli may be present in organs lin may not be effective in deep-seated streptococcal infections,
such as the lymph nodes, renal glomeruli, adrenal glands, brain, because stationary-phase organisms do not express PBPs. This
and splenic sinusoids.55,56 tolerance to penicillin has also been observed in group G strep-
tococcal infections.58 Approximately 10% to 15% of contem-
Treatment and Prognosis porary S. canis isolates from dogs in Europe have demonstrated
clindamycin resistance.59,60 Given this finding, the same combi-
In general, streptococcal infections should be treated with a nation could be used to treat NF in dogs and cats.
β-lactam drug, preferably a penicillin or a cephalosporin (Table
34-2). Early diagnosis and prompt treatment with appropriate Meningitis/Meningoencephalitis
antimicrobial drugs is essential to prevent more severe compli- Although high-dose penicillin has been used to treat strepto-
cations such as NF or STSS. In animals that present with severe coccal meningitis in human patients, penicillins typically have
signs such as septic shock or life-threatening pneumonia, initial limited penetration of the CSF. Trimethoprim-sulfamethoxazole
treatment should be with a broad-spectrum antimicrobial drug was used with apparent success to treat some dogs and cats with
combination, such as a β-lactam and an aminoglycoside, which streptococcal meningitis.21,61 See Chapter 90 for additional
CHAPTER 34  Streptococcal and Enterococcal Infections 341

TABLE 34-2
Suggested Antimicrobial Drugs for Treatment of Streptococcal Infections in Dogs and Cats
Drug Dose (mg/kg) Interval (hours) Route
Penicillin G potassium or sodium* 20,000-40,000 U/kg 6-8 IV, IM
Ampicillin sodium* 10-20 6-8 IV, IM, SC
Amoxicillin 6.6-20 8-12 PO
Cefazolin sodium 20-35 8 IV, IM
Cephalexin 10-30 (dogs), 15-20 (cats) 12 PO
Trimethoprim-sulfamethoxazole† 30 12 PO, IV

*Consider use with clindamycin, 10 mg/kg q12h IV or IM for necrotizing fasciitis or myositis; consider addition of gentamicin sulfate, 9-14 mg/kg
q24h IV, IM, or SC for dogs with S. equi subsp. zooepidemicus pneumonia because of synergy of this combination for group C streptococci.
†Consider use for treatment of streptococcal meningitis.

TABLE 34-3
Suggested Medications for Treatment of Enterococcal Infections in Dogs and Cats
Drug Dose (mg/kg) Interval (hours) Route
Ampicillin sodium* 10-20 6-8 IV, IM, SC
Amoxicillin* 6.6-20 8-12 PO
Vancomycin† 15 8 IV
Linezolid† 10 (dogs) 8 PO
Chloramphenicol 40-50 (dogs), 12.5-20 (cats) 6-8 (dogs), 12 (cats) PO, IV, IM
Nitrofurantoin‡ 2-3 8 PO

*In human patients, used with gentamicin sulfate, 1 mg/kg q12h IV for systemic enterococcal infections caused by enterococci susceptible to high-
level aminoglycosides, because of the synergy of this combination
†For enterococci that show high-level aminoglycoside resistance or when aminoglycosides cannot be used because of toxicity (animals with renal

failure). Use in veterinary patients is controversial. Refer to Chapter 8 for additional information on the use of vancomycin.
‡For resistant UTIs

information on treatment of bacterial meningitis. Ceftriaxone such as endocarditis or bacteremia. Enterococci are usually
has been used as an alternative to benzylpenicillin for treatment resistant to aminoglycosides at dilutions used in routine broth
of human streptococcal meningitis and attains higher concen- microdilution methods (low-level resistance). When the synergis-
trations in the CSF. tic combination is indicated, but low-level resistance is present,
isolates should be tested for high-level resistance (HLR) to ami-
Bacteremia and Endocarditis noglycosides. Aminoglycoside HLR results from production of
The reader is referred to Chapter 86 for information on treat- an aminoglycoside-modifying enzyme by enterococci. It emerged
ment of bacteremia and endocarditis. in the 1980s and has been a growing problem in both E. faecium
and E. faecalis isolates from human patients. Aminoglycoside
Enterococcal Infections HLR has also been reported in E. faecium and E. faecalis iso-
Suggested medications for treatment of enterococcal infections in lated from dogs and cats,36,38,62 but most isolates are susceptible
dogs and cats are shown in Table 34-3. Successful treatment of to high concentrations of gentamicin. In human patients, a com-
enterococcal infections requires knowledge of the intrinsic resis- bination of ceftriaxone and ampicillin has been recommended to
tance of enterococci to low levels of β-lactams and aminoglyco- treat endocarditis caused by aminoglycoside HLR enterococci.39
sides, the synergistic effect of high doses of these drugs, the lack When enterococci show penicillin resistance, vancomycin can be
of efficacy of trimethoprim-sulfonamide combinations in  vivo, substituted for penicillin, but this requires prolonged hospitaliza-
and the ability of enterococci to acquire resistance genes to a tion for intravenous administration. Single-agent treatment with
large number of other antimicrobial compounds. Aminopenicil- linezolid (see Chapter 8) is an oral alternative for treatment of
lins such as ampicillin generally have more potent activity against serious penicillin-resistant enterococcal infections, but in human
enterococci than does penicillin or the carbapenems. Bactericidal patients it is recommended that this only be used when combi-
regimens, which consist of a β-lactam and gentamicin (or strep- nations of β-lactams and gentamicin cannot be used because of
tomycin), are used in human patients to treat systemic infections resistance, toxicity, or treatment failure.39 Although most strains
342 SECTION 2  Bacterial Diseases

of enterococci isolated from humans are susceptible to line- close contact, and chronic subclinical carriage of infection in
zolid, resistant strains have appeared.63 Chloramphenicol may the oropharynx occurs in some individuals. Although family
be successful for treatment of resistant enterococcal bloodstream pets have been implicated as a reservoir in households with
infections when administered orally, but it is bacteriostatic, and recurrent streptococcal pharyngitis,66,67 colonization or infec-
concerns regarding adverse effects exist (see Chapter 8). tion of dogs and cats with GAS appears to be rare or nonexis-
Treatment of enterococcal UTIs with β-lactams such as tent when accurate streptococcal identification methods have
ampicillin or amoxicillin may be successful even when resis- been performed. One study found no evidence of oropharyn-
tance is documented in the susceptibility panel, because of the geal colonization by GAS in pets that were in contact with
high concentrations that these drugs achieve in the urine.39 children with acute pharyngitis.68 Even among human family
Nitrofurantoin has also been useful for treatment of resistant members who are in contact with children with streptococcal
enterococcal UTIs in humans, and routine treatment of subclini- pharyngitis, routine culture of asymptomatic individuals is not
cal bacteriuria caused by MDR enterococci is not recommended recommended, because penicillin fails to eradicate streptococ-
(see Chapter 89).64 When enterococci are present with other cal pharyngeal carriage.57
bacteria, especially in the urinary tract, treatment of the nonen- The α-hemolytic streptococcus S. pneumoniae is a commensal
terococcal pathogen may be sufficient to resolve the infection. of the human respiratory tract and can invade opportunistically
to cause a variety of clinical manifestations that include otitis
Prevention media, sinusitis, meningitis, bacteremia, and pneumonia. It pos-
sesses no Lancefield group antigens. Although extremely rare in
Because outbreaks of streptococcal infections in dogs and cats dogs and cats, infections with S. pneumoniae have been reported
have been associated with overcrowding, co-infections, and in a dog with meningoencephalitis,61 a cat with arthritis,69 and a
other stressors, appropriate shelter and kennel management and cat with cellulitis.70 It is likely that in these reports, the dog and
hygiene, as well as vaccination against other respiratory patho- cats acquired the infection from humans, because humans are the
gens, may help to prevent streptococcal infections. Proper wound natural hosts for S. pneumoniae.
care may prevent serious streptococcal infections such as bactere- Reports of human infection with S. canis and S. equi subsp.
mia, endocarditis, NF, and STSS. Methods to reduce nosocomial zooepidemicus exist. S. canis has increasingly been recognized as a
infections by enterococci are outlined in Chapter 11. Resistant cause of soft tissue infections in dog owners and has been isolated
enterococcal infections may be prevented through restricted and from humans with bacteremia, UTIs, osteomyelitis, and pneumo-
appropriate use of antimicrobial drugs and proper management nia.71,72 Rarely, S. equi subsp. zooepidemicus has been isolated
of underlying disorders that predispose to bacterial infections. from humans with meningitis, bacteremia, endocarditis, pneu-
monia, osteomyelitis, septic arthritis, acute nephritis, or STSS.
Public Health Aspects Most human infections with S. equi subsp. zooepidemicus have
been traced to an animal source, such as contact with horses, or
Streptococci include some of the most important bacterial consumption of inadequately pasteurized milk products or pork.
pathogens of humans. The β-hemolytic group A streptococ- However, apparent transmission of S. equi subsp. zooepidemicus
cus (GAS) S. pyogenes causes streptococcal pharyngitis (“strep from a dog with S. equi subsp. zooepidemicus pneumonia to a han-
throat”) and scarlet fever, as well as poststreptococcal rheu- dler, who developed severe systemic illness, has been described.73
matic fever, rheumatic heart disease, and glomerulonephritis. Dogs and cats are also a potential source for human infection
It has been estimated that globally, GAS cause 616 million by MDR enterococci. In one study, dogs that left a veterinary
new cases of pharyngitis each year.65 GAS has also been intensive care unit were colonized with MDR enterococci with
associated with other disease manifestations, which include the capacity for biofilm formation and horizontal transfer of
NF, STSS, myositis, vaginitis, and bacteremia. As a result, antimicrobial resistance genes.74
S. pyogenes is one of the most extensively studied of all strep- Because of the potential for transmission of streptococci and
tococci. There are more than 100 different serotypes, which enterococci between dogs, cats, and humans, preventative mea-
are classified based on the sequence of the M protein gene sures such as hand washing after pets are handled, household
(known as emm typing). M protein is a filamentous macromol- disinfection, and avoidance of close contact (such as allowing
ecule that traverses the cell wall and is a key virulence factor of pets to lick wounds or mucous membranes) should be consid-
GAS. Transmission between humans primarily occurs through ered, especially for immunocompromised pet owners.

CASE EXAMPLE the thoracolumbar region that first appeared 7 months


previously, enlarged to 6 × 4 cm in diameter over 2 months,
and then remained static in size for the subsequent 5 months.
Signalment: “Jazz” a 6-year-old female spayed miniature Skin scrapings and fungal culture were performed, and four
Pinscher from Fairfield in northern California skin biopsies were obtained. An underlying vasculitis was
History: Jazz was brought to the University of California, suspected, and treatment with vitamin E (200 U PO q12h)
Davis, Veterinary Medical Teaching Hospital for evaluation and an oral fatty acid supplement was recommended
of anorexia, lethargy, and diarrhea. Two days previously, pending the results of the diagnostic tests. The following
the dog had been seen by the dermatology service for a day the owner noted edema of the right pelvic limb and
focal area of alopecia and scaling on the dorsal midline in discontinued the medications. Over the next 24 hours, the
CHAPTER 34  Streptococcal and Enterococcal Infections 343

dog became lethargic, inappetent, and developed liquid Cholesterol 526 mg/dL (135-361 mg/dL)
diarrhea and polydipsia. Jazz had been vaccinated regularly Total bilirubin 0.5 mg/dL (0-0.2 mg/dL)
for canine distemper, adenovirus and parvovirus infections Urinalysis: SGr 1.021; pH 5.0, 1+ protein (SSA), 1+ bilirubin, 4+
and rabies and normally ate a commercial dry dog food. She hemoprotein, no glucose, no ketones, rare WBC/HPF, 0-2
was last vaccinated just after the skin lesion first appeared. RBC/HPF, few amorphous crystals, no bacteria seen
The dog was mostly indoors, going outside on leash walks Imaging Findings: Abdominal ultrasound: Enlargement of
only, and there were no other pets in the household. the uterine stump was present dorsal to the bladder. There
Other Medical History: Jazz had been spayed 5 weeks were no cystic structures within the stump and no evidence
previously, at which time pyometra was an incidental of fluid in this region or lymphadenopathy.
finding and was treated with a 7-day course of amoxicillin. Cytologic Findings: Needle aspiration of edema fluid from a
Physical Examination: region adjacent to the right axilla revealed large numbers of
Body Weight: 3.3 kg cocci, some in pairs, in the absence of significant numbers
General: Lethargic, alert and ambulatory, but weak. Vocalized of leukocytes.
on palpation of the thorax and abdomen. T = 102.2°F Microbiologic Testing: Aerobic and anaerobic blood
(39.0°C), HR = 120 beats/min, RR = 60 breaths/min, mucous culture (three isolator specimens): One colony of coagulase-
membranes pink, CRT = 1 s. negative staphylococci was grown on one plate. The other
Eyes, Ears, Nose, and Throat: Mild conjunctival hyperemia two plates were negative. No anaerobes were cultured.
and bilateral serous ocular discharge were present. The isolate was resistant to penicillin G, erythromycin, and
Integument: There was mild subcutaneous edema and a small trimethoprim-sulfamethoxazole, but susceptible to all other
amount of serosanguineous fluid in the region of the skin antimicrobials tested.
biopsies, but sutures remained intact. There was a single, Aerobic bacterial culture (aspirate of edema fluid): Large num-
raised erythematous lesion adjacent to the skin biopsies bers of Streptococcus canis colonies.
that measured 0.5 × 0.5 cm in diameter. Severe edema was Treatment and Outcome: Treatment with IV fluids (0.9%
noted in the cranioventral thoracic region, which extended NaCl with 20 mEq KCl/L) was initiated. Ampicillin (20 mg/kg IV
over an area that was 9 × 5 cm in diameter. q6h), enrofloxacin (5 mg/kg IV q24h), and butorphanol were
Other Systems, Including Peripheral Lymph Nodes: No administered. The dog’s body temperature subsequently
clinically significant findings. dropped to 98.0°F (36.7°C) and blood glucose concentration
Laboratory Findings: dropped to 30 mg/dL. Fluids were supplemented with
CBC: dextrose and a warming pad was used, and the dog’s
HCT 53.1% (40%-55%) temperature returned to normal limits. Within hours the
MCV 71.2 fL (65-75 fL) edema had spread to the abdomen, and evidence of
MCHC 36.7 g/dL (33-36 g/dL) bruising appeared on the right thorax and ventrum. The
Nucleated RBC 24/100 WBC raised erythematous lesion on the dorsum ulcerated, and
WBC 2200 cells/µL (6000-13,000 cells/µL) dehiscence of the biopsy site adjacent to this region became
Neutrophils 792 cells/µL (3000-10,500 cells/µL) apparent. Blood work showed a hematocrit of 39%, 2700
Band neutrophils 748 cells/µL white cells/µL, 1323 band neutrophils/µL, 702 neutrophils/µL,
Metamyelocytes 88 cells/µL 162 lymphocytes/µL, 513 monocytes/µL, and 37,000 platelets/
Myelocytes 44 cells/µL µL; serum sodium concentration was 142 mmol/L, potassium
Lymphocytes 308 cells/µL (1000-4000 cells/µL) 3.7 mmol/L, creatinine 0.4 mg/dL, BUN 23 mg/dL, albumin
Monocytes 220 cells/µL (150-1200 cells/µL) 1.2 g/dL, globulin 3.1 g/dL, cholesterol 221 mg/dL, and total
Platelets 97,000/µL (150,000-400,000 platelets/µL). bilirubin 2.3 mg/dL. The activities of ALT, AST, and ALP were 79
Markedly toxic band neutrophils, metamyelocytes, and U/L, 136 U/L, and 292 U/L, respectively. Treatment with dextran
myelocytes; moderately toxic neutrophils; and rare reac- and plasma was initiated, but systolic blood pressure dropped
tive lymphocytes were present. to 80 mm Hg, and the region adjacent to the biopsy sites
Serum Chemistry Profile: became firm and painful and tore when palpated. Ampicillin
Sodium 128 mmol/L (145-154 mmol/L) was substituted with penicillin G (40,000 U/kg IV q6h). Forty-
Potassium 4.4 mmol/L (3.6-5.3 mmol/L) eight hours after admission, the dog was anesthetized and a
Chloride 90 mmol/L (108-118 mmol/L) large portion of necrotic skin was surgically debrided from the
Bicarbonate 21 mmol/L (16-26 mmol/L) dog’s dorsum. The debrided region was covered with a stent.
Phosphorus 8.7 mg/dL (3.0-6.2 mg/dL) During recovery the dog was tachypneic, had increased lung
Calcium 10.0 mg/dL (9.7-11.5 mg/dl) sounds, and systolic blood pressure dropped to 50 mm Hg.
BUN 63 mg/dL ( 5-21 mg/dL) The dog developed respiratory arrest, and an endotracheal
Creatinine 0.9 mg/dL (0.3-1.2 mg/dL) tube was placed, from which bloody fluid emanated. Cardiac
Glucose 75 mg/dL (64-123 mg/dL) arrest then ensued, and there was no response to closed
Total protein 5.6 g/dL (5.4-7.6 g/dL) cardiopulmonary resuscitation efforts.
Albumin 2.2 g/dL (3.0-4.4 g/dL) Necropsy Findings: At necropsy, necrosis extended
Globulin 3.4 g/dL (1.8-3.9 g/dL) through the intercostal musculature of the right thoracic
ALT 185 U/L (19-67 U/L) wall. Histopathologic lesions included severe, focally
AST 128 U/L (19-42 U/L) extensive, hemorrhagic and necrotizing panniculitis and
ALP 539 U/L (21-170 U/L) fasciitis with intralesional bacteria and multifocal thrombi;

Continued
344 SECTION 2  Bacterial Diseases

diffuse necrotizing myositis of the right thoracic wall with perivascular neutrophilic dermatitis. No microorganisms were
intralesional bacteria; severe congestion and mild, multifocal seen, although only special staining with periodic acid–Schiff
hemorrhage and thrombosis within the kidneys; centrilobular (PAS) and Gomori’s methenamine silver (GMS) was performed.
congestion and severe bile stasis within the liver; congestion Diagnosis: NF, myositis, and STSS associated with
and hemorrhage within the lung; and generalized lymphoid Streptococcus canis infection
depletion and sinusoidal histiocytosis within lymph nodes. Comments: This rare case description illustrates how rapidly NF
Around the uterine stump, there was focal fibrosis and mild can progress and the ineffectiveness of antibiotic treatment
granulomatous serositis. Very small numbers of S. canis were in the presence of devitalized tissue. Clinical findings more
cultured from the uterine stump. Histopathology of the biopsy than satisfied the criteria for diagnosis of STSS in Box 34-1.
specimens that had been collected before the onset of systemic The source of infection and the role that the uterine stump
illness showed severe, diffuse adnexal atrophy; mild, chronic, infection played was unclear. Streptococcal bacteremia
lymphoplasmacytic mural folliculitis; focal granulomatous was not detected in this case. It is likely that the coagulase-
furunculosis with hyperkeratosis; and mild, multifocal, negative staphylococcus represented a contaminant.

SUGGESTED READINGS 14. DeWinter LM, Low DE, Prescott JF. Virulence of Streptococcus
canis from canine streptococcal toxic shock syndrome and necro-
Ghosh A, Dowd SE, Zurek L. Dogs leaving the ICU carry a very tizing fasciitis. Vet Microbiol. 1999;70:95-110.
large multi-drug resistant enterococcal population with capac- 15. Piva S, Zanoni RG, Specchi S, et  al. Chronic rhinitis due to
ity for biofilm formation and horizontal gene transfer. PLoS One. Streptococcus equi subspecies zooepidemicus in a dog. Vet Rec.
2011;6:e22451. 2010;167:177-178.
Lappin E, Ferguson AJ. Gram-positive toxic shock syndromes. Lancet 16. Acke E, Abbott Y, Pinilla M, et al. Isolation of Streptococcus zooep-
Infect Dis. 2009;9:281-290. idemicus from three dogs in close contact with horses. Vet Rec.
Prescott JF, DeWinter L. Canine streptococcal toxic shock syndrome 2010;167:102-103.
and necrotising fasciitis. Vet Rec. 1997;140:263. 17. Priestnall S, Erles K. Streptococcus zooepidemicus: an emerging
Priestnall S, Erles K. Streptococcus zooepidemicus: an emerging canine canine pathogen. Vet J. 2011;188:142-148.
pathogen. Vet J. 2011;188:142-148. 18. Sundberg JP, Hill D, Wyand DS, et al. Streptococcus zooepidemicus
as the cause of septicemia in racing greyhounds. Vet Med Small
REFERENCES Anim Clin. 1981;76:839-842.
1. Wilson LG. The early recognition of streptococci as causes of dis- 19. Britton AP, Davies JL. Rhinitis and meningitis in two shelter cats
ease. Med Hist. 1987;31(4):403-414. caused by Streptococcus equi subspecies zooepidemicus. J Comp
2. Schleifer KH, Kilpper-Balz R. Transfer of Streptococcus faecalis and Pathol. 2010;143:70-74.
Streptococcus faecium to the genus Enterococcus faecalis comb. 20. Blum S, Elad D, Zukin N, et  al. Outbreak of Streptococcus
nov. and Enterococcus faecium comb. nov. Int J Syst Bacteriol. equi subsp. zooepidemicus infections in cats. Vet Microbiol.
1984;34:31-34. 2010;144:236-239.
3. Lamm CG, Ferguson AC, Lehenbauer TW, et  al. Streptococcal 21. Martin-Vaquero P, da Costa RC, Daniels JB. Presumptive meningo-
infection in dogs: a retrospective study of 393 cases. Vet Pathol. encephalitis secondary to extension of otitis media/interna caused
2010;47:387-395. by Streptococcus equi subspecies zooepidemicus in a cat. J Feline
4. Tillman PC, Dodson ND, Indiveri M. Group G streptococcal epizo- Med Surg. 2011;13:606-609.
otic in a closed cat colony. J Clin Microbiol. 1982;16:1057-1060. 22. Larson LJ, Henningson J, Sharp P, et  al. Efficacy of the canine
5. Pesavento PA, Bannasch MJ, Bachmann R, et al. Fatal Streptococ- influenza virus H3N8 vaccine to decrease severity of clinical disease
cus canis infections in intensively housed shelter cats. Vet Pathol. after cochallenge with canine influenza virus and Streptococcus equi
2007;44:218-221. subsp. zooepidemicus. Clin Vaccine Immunol. 2011;18:559-564.
6. Taillefer M, Dunn M. Group G streptococcal toxic shock-like syn- 23. Priestnall SL, Erles K, Brooks HW, et al. Characterization of pneu-
drome in three cats. J Am Anim Hosp Assoc. 2004;40:418-422. monia due to Streptococcus equi subsp. zooepidemicus in dogs.
7. Kulendra E, Corr S. Necrotising fasciitis with sub-periosteal Strep- Clin Vaccine Immunol. 2010;17:1790-1796.
tococcus canis infection in two puppies. Vet Comp Orthop Trau- 24. Paillot R, Darby AC, Robinson C, et al. Identification of three novel
matol. 2008;21:474-477. superantigen-encoding genes in Streptococcus equi subsp. zooepi-
8. Sura R, Hinckley LS, Risatti GR, et  al. Fatal necrotising fasciitis demicus, szeF, szeN, and szeP. Infect Immun. 2010;78:4817-4827.
and myositis in a cat associated with Streptococcus canis. Vet Rec. 25. Vela AI, Falsen E, Simarro I, et  al. Neonatal mortality in puppies
2008;162:450-453. due to bacteremia by Streptococcus dysgalactiae subsp. dysgalactiae.
9. Prescott JF, DeWinter L. Canine streptococcal toxic shock syn- J Clin Microbiol. 2006;44:666-668.
drome and necrotising fasciitis. Vet Rec. 1997;140:263. 26. Devriese LA, Haesebrouck F. Streptococcus suis infections in
10. Lappin E, Ferguson AJ. Gram-positive toxic shock syndromes. horses and cats. Vet Rec. 1992;130:380.
Lancet Infect Dis. 2009;9:281-290. 27. Roels S, Devroye O, Buys H, et al. Isolation of Streptococcus suis
11. Iglauer F, Kunstyr I, Morstedt R, et al. Streptococcus canis arthritis from a cat with meningoencephalitis. Vet Microbiol. 2009;136:
in a cat breeding colony. J Exp Anim Sci. 1991;34:59-65. 206-207.
12. Kruger EF, Byrne BA, Pesavento P, et al. Relationship between clini- 28. Muckle A, Giles J, Lund L, et  al. Isolation of Streptococcus suis
cal manifestations and pulsed-field gel profiles of Streptococcus canis from the urine of a clinically ill dog. Can Vet J. 2010;51:773-774.
isolates from dogs and cats. Vet Microbiol. 2010;146:167-171. 29. Salasia SI, Lammler C, Devriese LA. Serotypes and putative viru-
13. Fulde M, Rohde M, Hitzmann A, et  al. SCM, a novel M-like lence markers of Streptococcus suis isolates from cats and dogs. Res
protein from Streptococcus canis, binds (mini)-plasminogen with Vet Sci. 1994;57:259-261.
high affinity and facilitates bacterial transmigration. Biochem J. 30. De Martino L, Nizza S, de Martinis C, et  al. Streptococcus
2011;434:523-535. ­constellatus-associated pyoderma in a dog. J Med Microbiol. 2011.
CHAPTER 34  Streptococcal and Enterococcal Infections 345

31. Schlegel L, Grimont F, Ageron E, et  al. Reappraisal of the tax- 49. Delgado M, Neto I, Correia JH, et  al. Antimicrobial resistance
onomy of the Streptococcus bovis/Streptococcus equinus complex and evaluation of susceptibility testing among pathogenic entero-
and related species: description of Streptococcus gallolyticus subsp. cocci isolated from dogs and cats. Int J Antimicrob Agents.
gallolyticus subsp. nov., S. gallolyticus subsp. macedonicus subsp. 2007;30:98-100.
nov. and S. gallolyticus subsp. pasteurianus subsp. nov. Int J Syst 50. Ossiprandi MC, Bottarelli E, Cattabiani F, et  al. Susceptibility
Evol Microbiol. 2003;53:631-645. to vancomycin and other antibiotics of 165 Enterococcus strains
32. Boleij A, van Gelder MM, Swinkels DW, et al. Clinical importance isolated from dogs in Italy. Comp Immunol Microbiol Infect Dis.
of Streptococcus gallolyticus infection among colorectal cancer 2008;31:1-9.
patients: systematic review and meta-analysis. Clin Infect Dis. 51. Naser SM, Vancanneyt M, De Graef E, et al. Enterococcus canin-
2011;53:870-878. testini sp. nov., from faecal samples of healthy dogs. Int J Syst Evol
33. Sykes JE, Kittleson MD, Pesavento PA, et al. Evaluation of the rela- Microbiol. 2005;55:2177-2182.
tionship between causative organisms and clinical characteristics of 52. Helie P, Higgins R. Diarrhea associated with Enterococcus faecium
infective endocarditis in dogs: 71 cases (1992-2005). J Am Vet Med in an adult cat. J Vet Diagn Invest. 1999;11:457-458.
Assoc. 2006;228:1723-1734. 53 Nicklas JL, Moisan P, Stone MR, et al. In situ molecular diagnosis
34. Tou SP, Adin DB, Castleman WL. Mitral valve endocarditis and histopathological characterization of enteroadherent Entero-
after dental prophylaxis in a dog. J Vet Intern Med. 2005;19: coccus hirae infection in pre-weaning-age kittens. J Clin Microbiol.
268-270. 2010;48:2814-2820.
35. Schleifer KH, Kilpper-Balz R. Transfer of Streptococcus faecalis 54. Rivera AM, Boucher HW. Current concepts in antimicrobial therapy
and Streptococcus faecium to the genus Enterococcus nom. rev. as against select gram-positive organisms: methicillin-resistant Staph-
Enterococcus faecalis com. nov. and Enterococcus faecium com. ylococcus aureus, penicillin-resistant pneumococci, and vancomy-
nov. Int J Syst Bacteriol. 1984;34:31-34. cin-resistant enterococci. Mayo Clin Proc. 2011;86:1230-1243.
36. Simjee S, White DG, McDermott PF, et al. Characterization of Tn1546 55. Garnett NL, Eydelloth RS, Swindle MM, et al. Hemorrhagic strep-
in vancomycin-resistant Enterococcus faecium isolated from canine tococcal pneumonia in newly procured research dogs. J Am Vet
urinary tract infections: evidence of gene exchange between human Med Assoc. 1982;181:1371-1374.
and animal enterococci. J Clin Microbiol. 2002;40:4659-4665. 56. Pesavento PA, Hurley KF, Bannasch MJ, et al. A clonal outbreak
37. Manson JM, Keis S, Smith JM, et  al. Characterization of a of acute fatal hemorrhagic pneumonia in intensively housed (shel-
­vancomycin-resistant Enterococcus faecalis (VREF) isolate from a ter) dogs caused by Streptococcus equi subsp. zooepidemicus. Vet
dog with mastitis: further evidence of a clonal lineage of VREF in Pathol. 2008;45:51-53.
New Zealand. J Clin Microbiol. 2003;41:3331-3333. 57. Bisno AL, Stevens DL. Streptococcus pyogenes. In: Mandell GL,
38. Abbott Y, Kirby BM, Karczmarczyk M, et  al. High-level genta- Bennett JE, Dolin R, eds. Principles and Practice of Infectious Dis-
micin-resistant and vancomycin-resistant Enterococcus faecium eases. 7th ed. Philadelphia, PA: Churchill Livingstone, Elsevier;
isolated from a wound in a dog. J Small Anim Pract. 2009;50: 2010:2593-2610.
194-197. 58. Pillai A, Thomas S, Williams C. Clindamycin in the treatment
39. Arias CA, Murray BE. Enterococcus species, Streptococcus bovis of group G beta-haemolytic streptococcal infections. J Infect.
group, and Leuconostoc species. In: Mandell GL, Bennett JE, Dolin 2005;51:e207-e211.
R, eds. Principles and Practice of Infectious Diseases. Philadelphia, 59. Lyskova P, Vydrzalova M, Mazurova J. Identification and antimi-
PA: Churchill Livingstone, Elsevier; 2010:2643-2653. crobial susceptibility of bacteria and yeasts isolated from healthy
40. Ghanem G, Hachem R, Jiang Y, et al. Outcomes for and risk fac- dogs and dogs with otitis externa. J Vet Med A Physiol Pathol Clin
tors associated with vancomycin-resistant Enterococcus faecalis Med. 2007;54:559-563.
and vancomycin-resistant Enterococcus faecium bacteremia in can- 60. Pedersen K, Jensen H, Finster K, et al. Occurrence of antimicrobial
cer patients. Infect Control Hosp Epidemiol. 2007;28:1054-1059. resistance in bacteria from diagnostic samples from dogs. J Antimi-
41. Mohamed JA, Huang DB. Biofilm formation by enterococci. J Med crob Chemother. 2007;60:775-781.
Microbiol. 2007;56:1581-1588. 61. Irwin PJ, Parry BW. Streptococcal meningoencephalitis in a dog.
42. Damborg P, Top J, Hendrickx AP, et  al. Dogs are a reservoir of J Am Anim Hosp Assoc. 1999;35:417-422.
ampicillin-resistant Enterococcus faecium lineages associated with 62. Harada T, Tsuji N, Otsuki K, et  al. Detection of the esp gene in
human infections. Appl Environ Microbiol. 2009;75:2360-2365. high-level gentamicin resistant Enterococcus faecalis strains from
43. Jackson CR, Fedorka-Cray PJ, Davis JA, et  al. Prevalence, spe- pet animals in Japan. Vet Microbiol. 2005;106:139-143.
cies distribution and antimicrobial resistance of enterococci iso- 63. Dowzicky MJ. Susceptibility to tigecycline and linezolid among
lated from dogs and cats in the United States. J Appl Microbiol. gram-positive isolates collected in the United States as part of the
2009;107:1269-1278. tigecycline evaluation and surveillance trial (TEST) between 2004
44. Simpson KW, Rishniw M, Bellosa M, et al. Influence of Enterococ- and 2009. Clin Ther. 2011;33:1964-1973.
cus faecium SF68 probiotic on giardiasis in dogs. J Vet Intern Med. 64. Swaminathan S, Alangaden GJ. Treatment of resistant enterococcal
2009;23:476-481. urinary tract infections. Curr Infect Dis Rep. 2010;12:455-464.
45. Bybee SN, Scorza AV, Lappin MR. Effect of the probiotic 65. Carapetis JR, Steer AC, Mulholland EK, et al. The global burden of
Enterococcus faecium SF68 on presence of diarrhea in cats and group A streptococcal diseases. Lancet Infect Dis. 2005;5:685-694.
dogs housed in an animal shelter. J Vet Intern Med. 2011;25: 66. Mayer G, Van Ore S. Recurrent pharyngitis in family of four.
856-860. Household pet as reservoir of group A streptococci. Postgrad Med.
46. Lappin MR, Veir JK, Satyaraj E, et  al. Pilot study to evalu- 1983;74:277-279.
ate the effect of oral supplementation of Enterococcus faecium 67. Falck G. Group A streptococci in household pets’ eyes—a source of
SF68 on cats with latent feline herpesvirus 1. J Feline Med Surg. infection in humans? Scand J Infect Dis. 1997;29:469-471.
2009;11:650-654. 68. Wilson KS, Maroney SA, Gander RM. The family pet as an
47. Veir JK, Knorr R, Cavadini C, et al. Effect of supplementation with unlikely source of group A beta-hemolytic streptococcal infection
Enterococcus faecium (SF68) on immune functions in cats. Vet in humans. Pediatr Infect Dis J. 1995;14:372-375.
Ther. 2007;8:229-238. 69. Stallings B, Ling GV, Lagenaur LA, et  al. Septicemia and septic
48. Benyacoub J, Czarnecki-Maulden GL, Cavadini C, et  al. Supple- arthritis caused by Streptococcus pneumoniae in a cat: possi-
mentation of food with Enterococcus faecium (SF68) stimulates ble transmission from a child. J Am Vet Med Assoc. 1987;191:
immune functions in young dogs. J Nutr. 2003;133:1158-1162. 703-704.
346 SECTION 2  Bacterial Diseases

70. Zhang S, Wilson F, Pace L. Streptococcus pneumoniae-associated 73. Abbott Y, Acke E, Khan S, et al. Zoonotic transmission of Strepto-
cellulitis in a two-month-old domestic shorthair kitten. J Vet Diagn coccus equi subsp. zooepidemicus from a dog to a handler. J Med
Invest. 2006;18:221-224. Microbiol. 2010;59:120-123.
71. Galperine T, Cazorla C, Blanchard E, et  al. Streptococcus canis 74. Ghosh A, Dowd SE, Zurek L. Dogs leaving the ICU carry a very
infections in humans: retrospective study of 54 patients. J Infect. large multi-drug resistant enterococcal population with capacity
2007;55:23-26. for biofilm formation and horizontal gene transfer. PLoS One.
72. Lam MM, Clarridge 3rd JE, Young EJ, et al. The other group G 2011;6:e22451.
Streptococcus: increased detection of Streptococcus canis ulcer
infections in dog owners. J Clin Microbiol. 2007;45:2327-2329.
CHAPTER 35

Staphylococcus Infections
Jane E. Sykes

infect dogs and cats are Staphylococcus pseudintermedius (pre-


Overview of Staphylococcal Infections viously identified as Staphylococcus intermedius) and Staphylo-
in Dogs and Cats coccus aureus. Staphylococcus schleiferi has also been isolated
First Described: 1882 in Scotland (Alexander Ogsten)1 from dogs (and rarely cats) with pyoderma and otitis externa
and has been divided into S. schleiferi subsp. schleiferi, which
Causes: In dogs and cats, the most common causes are Staph-
is coagulase-negative, and S. schleiferi subsp. coagulans, which
ylococcus pseudintermedius, Staphylococcus schleiferi,
is coagulase-positive. However, these may instead represent a
Staphylococcus aureus, and various coagulase-negative
single species, S. schleiferi, with variable coagulase production.3
staphylococci, which are gram-positive coccoid bacteria.
Coagulase-negative staphylococci (CoNS) tend to be of lower
Geographic Distribution: Worldwide virulence than coagulase-positive staphylococci but occasion-
Mode of Transmission: Direct contact (or through fomi- ally cause disease in immunocompromised hosts. Examples of
tes). Organisms are often commensals that invade coagulase-negative staphylococci are Staphylococcus felis and
opportunistically. Staphylococcus epidermidis. S. felis has been recognized as a
urinary tract pathogen in cats.4 CoNS are often not identified to
Major Clinical Signs: Pyoderma, otitis externa, surgical site
the species level by microbiology laboratories.
infections, osteomyelitis, bacteremia and endocarditis,
In recent years, the prevalence of antimicrobial drug resistance
bronchopneumonia, urinary tract infections, ocular surface
has increased in Staphylococcus isolates obtained from canine and
infections, rarely necrotizing fasciitis and toxic shock syn-
feline infections. A significant proportion of resistant isolates pos-
drome, pyothorax and peritonitis, discospondylitis, arthritis
sess the mecA gene, which encodes an altered penicillin binding
Differential Diagnoses: Primarily other gram-positive and gram-­ protein (PBP), known as PBP2a. The mecA gene is located on a
negative bacterial infections large genetic element, the staphylococcal cassette chromosome.
Human Health Significance: Humans can become colonized This PBP has low affinity for all β-lactam drugs (penicillins, cepha-
with staphylococcal isolates that colonize or infect dogs losporins, and carbapenems). As a result, staphylococci that possess
and cats. Methicillin-resistant S. aureus, which can persis- this gene are also resistant to the penicillinase-resistant penicillins
tently colonize humans, is of greatest concern, although (oxacillin and methicillin), and so are termed methicillin-resistant
colonization of dogs with MRSA appears to be transient. Staphylococcus (MRS) species. Many MRS also possess resistance
S. pseudintermedius, which persistently colonizes dogs, has genes to other antimicrobial drug classes. There is no evidence that
occasionally been isolated from sick and healthy people MRS are more virulent than methicillin-susceptible staphylococci,
who are in contact with dogs, and is of concern because of but infections with MRS are more difficult to treat with antimicro-
its tendency to show a high degree of multidrug resistance. bial drugs. The prevalence of MRS isolated from dogs has ranged
from 0.6% to as high as 67%, depending on the population sam-
pled (healthy versus disease, hospitalized versus outpatients), geo-
graphic location, study dates, and sites of specimen collection. Of
Etiology and Epidemiology 89 staphylococcal isolates from dogs with superficial pyoderma
seen in 2010 and 2011 by the University of California, Davis, Vet-
Staphylococcus spp. are gram positive, nonmotile, non–spore erinary Dermatology service, 38.2% were MRS. This compared
forming, and usually catalase-positive cocci, which occur sin- with 27.3% of 33 isolates from a primary care clinic in the same
gly and in pairs or grapelike clusters (Figure 35-1). Most spe- region.5 Methicillin-resistant S. pseudintermedius (MRSP) isolates
cies are facultative anaerobes. Staphylococci colonize the skin are generally resistant to more antimicrobial drug classes than
and mucous membranes of an enormous variety of animal spe- methicillin-resistant S. aureus (MRSA) isolates from dogs and cats.
cies, and disease generally follows a breakdown in normal host Severe multidrug resistance is also prevalent among CoNS.
defenses. Staphylococci can also contaminate surfaces in the
environment, where they may survive for several months. Staphylococcus pseudintermedius
Staphylococci are classified as coagulase negative or coag- S. pseudintermedius is the most common Staphylococcus species
ulase positive based on their ability to produce coagulase, an isolated from dogs. It is a commensal of the skin and mucous
enzyme that cleaves fibrinogen into fibrin and results in coagu- membranes of dogs and cats and possibly one of the most com-
lation of plasma. There are more than 40 species in the genus mon bacterial pathogens treated by veterinarians. It primar-
Staphylococcus,2 many of which are adapted to live on certain ily colonizes the anal mucosa and nares of healthy dogs and
host species. The major coagulase-positive staphylococci that cats, but it can also be isolated from the mouth, forehead, and

347
348 SECTION 2  Bacterial Diseases

Before 2005, S. pseudintermedius were identified as S. inter-


medius. The name S. pseudintermedius was defined when isolates
from a dog, cat, horse, and parrot were analyzed using molecu-
lar methods.6 Subsequently, a molecular analysis of organisms
from dogs and cats that had been previously identified as S. inter-
medius revealed that they were actually S. pseudintermedius.
S. pseudintermedius belongs to the “S. intermedius group” (SIG),
which comprises the three closely related species S. intermedius,
S. pseudintermedius, and S. delphini. It is difficult to differenti-
ate among these species with phenotypic methods alone; genetic
typing methods are required. Because genetic typing methods are
not available for routine identification in veterinary diagnostic
laboratories, isolates from dogs with phenotypic characteristics
typical of S. intermedius are now generally identified as S. pseud-
intermedius.7 However, some laboratories still describe strains
from dogs that have biochemical characteristics that do not quite
match that typical for the type strain of S. pseudintermedius as
“SIG organisms.”
The whole genome of S. pseudintermedius has been sequenced,
which is a major contribution to our understanding of the patho-
A genesis of infections caused by this organism.8 Like S. aureus,
S. pseudintermedius possesses numerous mobile genetic elements
and potential virulence factors, such as toxin genes that encode
for a superantigen, leukotoxin, hemolysins, and exfoliative tox-
ins, as well as an array of proteases.
Methicillin resistance has emerged as a serious and wide-
spread problem in S. pseudintermedius isolates from both
healthy and sick dogs and cats. Molecular analysis has shown
that among dogs, two distinct, major clones of MRSP have
spread across Europe and North America, respectively.9 Many
isolates of S. pseudintermedius not only are methicillin resistant,
but also acquire resistance genes to other classes of antimicrobi-
als, including tetracyclines, trimethoprim-sulfonamides, fluoro-
quinolones, chloramphenicol, and macrolides. Isolates that are
resistant to three or more different classes of antimicrobial drugs
are defined as multiple drug resistant (MDR) isolates. Currently,
many isolates remain susceptible to chloramphenicol and amino-
glycosides, but the prevalence of resistance to chloramphenicol
has increased among MRSP isolates. MDR S. pseudintermedius
isolates are no more virulent or difficult to disinfect than sus-
ceptible isolates, but they are more difficult to eliminate using
B antimicrobial drugs. Risk factors for MRSP infection identified
FIGURE 35-1  Histopathology of the integument from the left inguinal region and in dogs include previous hospitalization, living in an urban envi-
flank of an 8-year old male neutered boxer dog that had severe, regionally extensive acute ronment, older age, and, most consistently, previous antimicro-
neutrophilic cellulitis and panniculitis accompanied by septic shock. A methicillin suscep- bial drug treatment. Household contacts of dogs and cats with
tible Staphylococcus pseudintermedius was cultured from an aspirate of cellulitis fluid. MRSP infection are also frequently colonized with MRSP.10
The dog had also been treated with prednisone and azathioprine for 4 years to maintain
remission for immune-mediated hemolytic anemia and thrombocytopenia. A, H&E stain. Staphylococcus aureus
Large numbers of degenerate neutrophils are present. B, Staining with Brown and Benn
stain reveals numerous of gram-positive coccoid bacteria, individually, in pairs, and in clus- S. aureus colonizes the skin and mucous membranes of humans
ters.1000× oil magnification. and is one of the most common community-acquired and
­hospital-acquired bacterial pathogens of people. The anterior
nares and the throat are preferred niches for the organism,
inguinal region. It can colonize the skin transiently following which can be isolated from the anterior nares of up to 30%
grooming and licking in dogs with pruritis. Cats have a much of the healthy human population. Methicillin resistance is a
lower prevalence of colonization with S. pseudintermedius than growing problem among isolates from humans and animals.
dogs. S. pseudintermedius invades tissues opportunistically Both hospital-acquired methicillin-resistant (HA-MRSA) and
and is a major cause of superficial and deep pyoderma, otitis community-acquired methicillin-resistant (CA-MRSA) strains
externa, urinary tract infections (UTIs), and wound and surgical have been identified in infected human patients, and invasive
site infections, as well as bronchopneumonia, ocular infections, S. aureus infections are associated with high mortality rates.
bacteremia, osteomyelitis, and infections of body cavities. An When compared with S. pseudintermedius, S. aureus tends
underlying immunosuppressive condition or break in host bar- not to colonize dogs. For example, staphylococci were isolated
riers is present in the majority of infections. from 37 of 50 healthy dogs from Pennsylvania, and 6 (12%) of
CHAPTER 35  Staphylococcus Infections 349

the dogs were colonized with S. aureus; 68% were colonized with A number of enterotoxins have been identified in S. aureus and
S. pseudintermedius.11 MRSA strains that colonize dogs may be S. pseudintermedius; these can cause food poisoning in humans
identical to those that infect in-contact humans, but sometimes but have not been recognized as a cause of disease in dogs and
strains found on dogs differ from those found on in-contact cats. Staphylococcal enterotoxins are heat stable, so they are not
humans.12 The chance that a dog that is in contact with an inactivated by cooking, even when the bacteria themselves are
infected human will be colonized with MRSA decreases with the destroyed. Staphylococcal toxic shock syndrome results from
time after diagnosis of human MRSA infection, which suggests the expression of superantigens, which include toxic shock syn-
transient carriage of MRSA by dogs. MRSA was not transferred drome toxin 1, as well as a large number of enterotoxins and
between apparently healthy dogs in a kennel of dogs, and after exotoxins that can also act as superantigens. Cellulitis, necrotiz-
treatment of an MRSA wound infection in one dog, all dogs tested ing fasciitis, and toxic shock syndrome has been rarely reported
negative for MRSA within 2 weeks.13 Although staphylococci in dogs infected with S. pseudintermedius.16,17 Most toxic shock
are isolated less commonly from cats, colonization with S. aureus syndrome and necrotizing fasciitis cases in dogs and cats are
is more prevalent and certain S. aureus strains may be commen- instead caused by Streptococcus canis (see Chapter 34). Scalded
sals of cats; staphylococci were isolated from 17 of 50 healthy skin syndrome results from infection of humans with S. aureus
cats, 59% of which were colonized with S. aureus and 65% with strains that produce an exfoliative toxin, which hydrolyses the
S. pseudintermedius.14 intercellular glycoprotein desmoglein-1. Strains of S. pseudin-
In dogs and cats, S. aureus infection has been associated with termedius that possess exfoliative toxin genes have been isolated
clinical manifestations that include pyoderma, otitis externa, UTIs, more commonly from dogs with pyoderma than from healthy
surgical site infections, osteomyelitis, and bacteremia. Although dogs, so exfoliative toxin may also play a role in canine disease.
MRSA infections are uncommonly diagnosed in dogs and cats, Panton-Valentine toxin (PVL) is a leukocidin that is found in
they have been increasingly recognized over the past 2 decades. CA-MRSA strains but only a small percentage of HA-MRSA
When canine infections with MRSA do occur, they are often of and MSSA strains, and strains that encode PVL have been asso-
human hospital origin, but human community-acquired strains ciated with skin and soft tissue infections and hemorrhagic
have also been isolated. In one study, strong risk factors for MRSA pneumonia in children and young adults. Although a similar
infection (as opposed to methicillin-susceptible S. aureus [MSSA] toxin has been identified in S. pseudintermedius, the role of PVL
infection) were the number of antimicrobial courses administered, in the pathogenesis of staphylococcal infections in dogs and cats
the number of days admitted to veterinary clinics, and a history is unknown.
of surgical implant placement.15 There has been concern that the Some strains of S. pseudintermedius and S. aureus produce
use of certain antibiotics, such as cephalosporins and fluoroquino- biofilm, which may contribute to the ability of these organisms
lones, may select for MRSA infections in dogs. to persist in the environment, cause nosocomial infections, and
chronic recurrent infections that respond poorly to antimicro-
Staphylococcus schleiferi bial treatment.18
Coagulase-positive and coagulase-negative S. schleiferi have
primarily been isolated from dogs with superficial pyoderma Diagnosis
and otitis externa, as well as from the ear canals of healthy
dogs. Rarely, UTIs and respiratory infections with S. schleiferi Laboratory Abnormalities
have been described in dogs.3 In dogs with pyoderma and otitis Complete Blood Count, Biochemistry, and Urinalysis Findings
externa, S. schleiferi is not as prevalent as S. pseudintermedius Dogs with staphylococcal infections of the skin and ear canal
but in most studies has been isolated as often, or more often, often have no clinically significant hematologic or biochemical
than S. aureus. Coagulase-negative S. schleiferi is a commensal findings. Systemic infections may be associated with neutrophilia
of the human axilla but can cause disease in immunosuppressed and a left shift, toxic neutrophils, lymphopenia, and monocyto-
humans. Coagulase-positive S. schleiferi may be specialized to sis. The urinalysis of dogs and cats with staphylococcal UTIs
colonize dogs. may reveal pyuria, proteinuria, or hematuria, and sometimes
S. schleiferi isolates from dogs with skin disease are often cocci in clusters may be evident on sediment examination.
methicillin-resistant as a result of mecA gene carriage. In a
study of 225 S. schleiferi isolates from dogs from the eastern Cytologic Abnormalities
United States, of which 52% were coagulase negative and 42% Aspirates of affected tissues or pus or respiratory lavage fluid
were coagulase positive, methicillin resistance was identified in specimens from animals with staphylococcal infections may
57% of the isolates.3 Methicillin resistance was more prevalent contain large numbers of degenerate and nondegenerate neutro-
among coagulase-negative isolates. Isolates from dogs with pyo- phils. The presence of cocci in clusters suggests staphylococcal
derma may be more likely to be methicillin resistant than those infection.
from dogs with otitis externa.
Microbiological Tests
Clinical Features Isolation and Identification
Staphylococci that infect dogs and cats are readily isolated on
Signs and Their Pathophysiology routine bacteriologic media from aspirates, tissue biopsies, body
Staphylococci can cause pyoderma, otitis externa, pneumonia, fluid, and lavage specimens. Colonies typically appear within
UTIs, soft tissue infections, surgical site infections, bacteremia, 24 hours. Staphylococcal species are identified on the basis
ocular infections, and endocarditis in dogs and cats. of phenotypic and biochemical reactions such as production
S. aureus and S. pseudintermedius possess several adhes- of coagulase and/or clumping factor, production of hemoly-
ins and produce an array of toxins. The clinical effect of these sins, ability to ferment carbohydrates, and acetoin production
toxins is best characterized for S. aureus infections in humans. (Vogues-Proskauer reaction). PCR and sequencing of ribosomal
350 SECTION 2  Bacterial Diseases

RNA genes (16S or 23S) may be necessary for identification referred to Chapters 84 to 90 for treatment recommendations
of some species. CoNS are often not identified to the species for different organ systems. Identification and management
level because molecular techniques are often required for accu- of the underlying cause (rather than repeated treatment with
rate identification. Because CoNS are of low pathogenicity, antimicrobial drugs) is of paramount importance to minimize
they may be contaminants, especially if they are isolated from selection for resistant organisms. Any foreign material (such as
nonsterile sites, growth is minimal (1+), or another pathogen catheters, implants) must be removed. Bathing and topical treat-
is present. However, they may occasionally be pathogenic. In ments should be considered for pyoderma as an alternative to
particular, isolation of S. felis from urine or coagulase-negative antimicrobial drug therapy (see Chapter 84). The use of topi-
S. schleiferi from dogs with pyoderma or otitis may have clinical cal wound treatments such as accelerated hydrogen peroxide,
significance. chlorhexidine, or silver sulfadiazine could also be considered
Evaluation of antimicrobial susceptibility is of critical impor- provided infection is superficial.
tance because of the high prevalence of methicillin-resistant and When systemic antimicrobial treatment is necessary, staphy-
multidrug-resistant staphylococci in some parts of the world. lococcal infections should ideally be treated based on the results
Susceptibility testing for staphylococci should be performed of culture and susceptibility testing and/or knowledge of the
according to recommendations of groups such as the Clini- regional prevalence of methicillin resistance and multidrug resis-
cal and Laboratory Standards Institute (CLSI). Oxacillin and tance, because this can vary, not only between countries but also
cefoxitin are used in the laboratory as a surrogate for detection between different states or regions within a state. In regions where
of methicillin resistance. Laboratories should report methicillin- methicillin resistance is rare, a β-lactam drug such as cephalexin,
resistant staphylococci as resistant to all other penicillins, car- clavulanic acid–amoxicillin, or a penicillinase-resistant penicillin
bapenems, cephalosporins, and β-lactam/β-lactamase inhibitor could be used empirically (Table 35-1), and culture and suscepti-
combinations, regardless of the results of in vitro susceptibility bility testing may not be necessary for uncomplicated infections
test results for these agents. such as newly diagnosed pyoderma or UTIs.
Where methicillin resistance is prevalent in the region, culture
Molecular Diagnosis Using the Polymerase Chain Reaction and susceptibility testing is recommended. If this is not possible,
Real-time PCR assays have been developed that detect and dif- a reasonable first choice for uncomplicated infections that are
ferentiate some Staphylococcus species, as well as identify the not life threatening is clindamycin. Clindamycin is an attractive
presence of the mecA gene. These assays are available in some choice given that many infections are skin, orthopedic, or soft
laboratories for diagnosis of MRSA infections in humans, but tissue infections (sites to which clindamycin distributes well);
to date have only been used on a research basis to characterize it is inexpensive, inhibits toxin production by staphylococci,
staphylococci isolated from dogs and cats. and is available in oral and intravenous formulations. However,
some MRS demonstrate inducible clindamycin resistance, a
Treatment and Prognosis phenomenon whereby the organism is susceptible to clindamy-
cin in vitro but rapidly develops resistance to clindamycin when
The approach to treatment of staphylococcal infections depends exposed to macrolides (such as erythromycin) or lincosamides
on the site of infection and the severity of disease; the reader is in vivo, owing to bacterial methylation of the ribosomal binding

TABLE 35-1
Suggested Antimicrobial Drugs for Treatment of Staphylococcal Infections in Dogs and Cats
Predicted/Determined
Staphylococcal
Susceptibility Drug Dose (mg/kg) Interval (hours) Route
Methicillin-susceptible Ampicillin sodium 10-20 6-8 IV, IM, SC
Cefazolin sodium 20-35 8 IV, IM
Cephalexin 10-30 (dogs), 15-20 (cats) 12 PO
Methicillin-resistant* Trimethoprim-sulfamethoxazole 30 12 PO, IV
Clindamycin† 11 12 PO
Doxycycline 10 12 PO
Rifampin 5-10 12 PO
Amikacin 15-30 (dogs), 10-14 (cats) 24 IV, IM, SC
Vancomycin‡ 15 6-8 (dogs), 8 (cats) IV
Linezolid‡ 10 (dogs) 8 PO, IV
Nitrofurantoin (macrocrystalline 2-3 8 PO
formulation)§

*Susceptibility testing recommended if methicillin resistance is likely based on regional or hospital prevalence data. Many isolates are resistant to
clindamycin, trimethoprim-sulfamethoxazole, doxycycline; some are resistant to chloramphenicol. Consider topical antibacterial treatment if possible.
†May not be effective if erythromycin resistance is present on the susceptibility panel due to inducible clindamycin resistance.
‡Reserve for serious infections that are resistant to all other reasonable alternatives, on the basis of culture and susceptibility testing. See Table 8-6

for vancomycin administration protocol.


§For resistant urinary tract infections only.
CHAPTER 35  Staphylococcus Infections 351

site for clindamycin.19 The end result is treatment failure or resistant to trimethoprim-sulfamethoxazole, doxycycline, and
relapse, although some treatment successes have been reported chloramphenicol, and toxicity may limit the usefulness of trim-
in human patients infected with S. aureus strains that have ethoprim-sulfamethoxazole and chloramphenicol (see Chapter
inducible clindamycin resistance. Staphylococci with inducible 8). Aminoglycosides must be given parenterally and have the
clindamycin resistance are resistant to macrolides such as eryth- potential to cause nephrotoxicity, and their activity is reduced
romycin and susceptible to clindamycin in routine susceptibil- in the presence of pus. The use of rifampin alone is contro-
ity tests. Nevertheless, not all staphylococci with this resistance versial, because rifampin monotherapy has been associated
pattern demonstrate inducible clindamycin resistance. Identifi- with development of resistance in S. aureus infections.23 As a
cation of inducible clindamycin resistance requires a special test result, the use of rifampin in combination with other drugs has
known as a D-zone test. It involves culture of the organism on been recommended. Minocycline has received attention as an
agar in the presence of an erythromycin and clindamycin disk. alternative to doxycycline because some doxycycline-resistant
Organisms near to the erythromycin disk express enhanced strains (strains that possess the tetracycline efflux protein tetK)
resistance to clindamycin, which results in a D shape to the remain susceptible to minocycline. In contrast, strains that pos-
zone of inhibition around the clindamycin disk (Figure 35-2). sess tetM (which protects the ribosome from tetracycline bind-
At the time of writing, inducible clindamycin resistance appears ing) are resistant to all tetracyclines. Additional information on
to be prevalent in S. aureus isolates from dogs and cats (18% the pharmacokinetics and definition of clinical breakpoints for
in one study of 62 isolates) but is rarely reported in S. pseud- minocycline are required.
intermedius (0 of 46 isolates in one study, and 1 of 60 isolates Vancomycin, tigecycline, daptomycin, quinupristin-
in another study).20,21 As a result, it is reasonable to assume dalfopristin, and linezolid are important drugs for MRSA
that disease caused by S. pseudintermedius isolates that are sus- infections in humans. Whether these drugs should be used to
ceptible to clindamycin on routine susceptibility testing is likely treat companion animals has been debated, given that they are
to respond to treatment with clindamycin. However, in some already widely used in humans and that they may be required
geographic locations, a high percentage of canine MRSP isolates to save the lives of companion animals. Cost and/or the need
are overtly resistant to clindamycin, which suggests that clinda- for parenteral administration (vancomycin, tigecycline, dapto-
mycin may not be an ideal empiric treatment choice in these mycin, and quinupristin-dalfopristin) can also limit the useful-
locations. The use of fluoroquinolones to treat staphylococcal ness of these drugs in companion animals. At a minimum, their
infections has been controversial, because they have been asso- use should be reserved for situations when no other treatment
ciated with treatment failures and development of methicillin alternatives exist and when the infection is life threatening but
resistance in human S. aureus infections. In one study, a fluoro- has the potential to be resolved with appropriate treatment. The
quinolone control program in a tertiary human hospital led to a use of vancomycin or linezolid may be indicated in dogs with
significant reduction in fluoroquinolone-resistant Pseudomonas bacteremia or endocarditis due to MRSA, MRSP, or methicillin-
aeruginosa and MRSA rates over 4 years.22 resistant CoNS that are resistant to all options other than amino-
If severe and invasive infection with an MRS is a possibil- glycosides and concerns for aminoglycoside toxicity exist.
ity, a combination of clindamycin and an aminoglycoside
could be administered parenterally, pending the results of cul- Prevention
ture and susceptibility testing. Antimicrobials that could be
considered for life-threatening methicillin-resistant and multi- Prevention of staphylococcal infections involves attention to
drug-resistant staphylococcal infections include trimethoprim- the underlying cause of disease (such as allergic dermatitis), and
sulfamethoxazole, doxycycline, aminoglycosides, rifampin, strict veterinary clinic and veterinary hospital infection control
chloramphenicol, vancomycin, tigecycline, quinupristin-dal- practices (see Chapter 11). Because canine MRSA infections
fopristin, and linezolid (see Chapter 8). Some MRS are also may be acquired from colonized or infected humans, trans-
mission to dogs may be reduced by hand washing and general
surface cleaning and disinfection in the home environment (see
Public Health Aspects, next).

Public Health Aspects


Most concern regarding the public health risk of staphylococcal
infection or colonization in companion animals has related to
MRSA. However, because colonization of dogs with MRSA is
transient, other humans are a more significant source of human
MRSA infection. Nevertheless, some studies suggest that veteri-
narians who work with small animals may be at increased risk for
colonization with MRSA. Although around 30% of the human
population is colonized with S. aureus, the prevalence of MRSA
colonization is generally much lower than this, less than 2%. In
Australia, dog and cat veterinarians had a fivefold higher risk for
MRSA carriage when compared with industry and government
FIGURE 35-2  Positive D-zone test for inducible clindamycin resistance in staphylo- veterinarians.24 Small animal veterinarians in the United King-
cocci. Organisms exposed to erythromycin express enhanced resistance to clindamycin, dom were also at increased risk of colonization.25 In a study from
which leads to distortion of the zone of inhibition around the clindamycin disk. (Courtesy the United States, 17.3% of veterinarians and veterinary techni-
Dr. Scott Weese, University of Guelph.) cians were colonized, and the rate of colonization did not differ
352 SECTION 2  Bacterial Diseases

between small- and large-animal veterinarians.26 In another study, Routine screening and decontamination is not recommended
only large-animal practice was associated with colonization.27 because dogs appear to resolve infection over time. Topical
Regardless of the direction of transmission (human to pet mupirocin, which has been used for decontamination in humans,
versus pet to human), owners of dogs and cats diagnosed with is not likely to be effective in dogs and cats given the large size of
MRSA infection should be educated about the need to wash their nasal cavities, and concerns have been raised regarding the
their hands after handling their pets, clean and disinfect surfaces need for and efficacy of decolonization with systemic antibiot-
that might become contaminated, and not let their pet lick their ics, which may only select for antimicrobial resistance.
face or open wounds. Infected sites should be covered with a Human infections with S. pseudintermedius occur occasion-
suitable dressing to minimize contamination of the environment ally in association with pet contact, although the organism
whenever possible. Owners should be instructed to talk to their generally tends not to colonize humans. However, coloniza-
health care providers if they have concerns about their health or tion with MRSP was reported more frequently in small-animal
the health of others in their household. In the hospital environ- dermatologists than colonization with MRSA.28,29 Precautions
ment, transmission of MRSA may be prevented through the use recommended for MRSA infection are also indicated for MRSP
of a strict hospital infection control program (see Chapter 11). infections.

CASE EXAMPLE Lymph Nodes: All lymph nodes were normal in size.
Laboratory Findings:
CBC:
Signalment: “Elliot,” a 10-year-old male neutered miniature HCT 32.2% (40%-55%)
schnauzer from San Francisco, CA MCV 60.3 fL (65-75 fL)
History: Elliot was seen for a possible pacemaker infection. MCHC 32.3 g/dL (33-36 g/dL)
One month after placement of a dual-chamber pacemaker Reticulocytes 9100 cells/µL (7000-65,000 cells/µL)
for sick sinus syndrome, a fluctuant mass was noted WBC 29,680 cells/µL (6000-13,000 cells/µL)
around the generator. The seroma was aspirated, and Neutrophils 22,854 cells/µL (3000-10,500 cells/µL)
cytologic examination of the aspirate showed many Lymphocytes 4155 cells/µL (1000-4000 cells/µL)
neutrophils. Treatment with clavulanic acid-amoxicillin Monocytes 594 cells/µL (150-1200 cells/µL)
was initiated (25 mg/kg PO q12h). One month later, there Eosinophils 890 cells/µL (0-1500 cells/µL)
had been no apparent improvement. The wound was Platelets 446,000 platelets/µL (150,000-400,000 platelets/µL)
flushed with chlorhexidine solution and treatment with Slight toxicity of neutrophils and band neutrophils.
chloramphenicol was initiated, but the problem persisted. Serum Chemistry Profile:
The chloramphenicol was discontinued, and the dog was Sodium 140 mmol/L (145-154 mmol/L)
referred for further evaluation. The owners reported no Potassium 5.6 mmol/L (3.6-5.3 mmol/L)
vomiting, diarrhea, or decreased appetite. Elliot had also Chloride 103 mmol/L (108-118 mmol/L)
been diagnosed with early myxomatous degeneration of Bicarbonate 16 mmol/L (16-26 mmol/L)
the mitral valve based on the results of echocardiography, Phosphorus 7.5 mg/dL (3.0-6.2 mg/dL)
associated with a grade III/VI left apical systolic murmur at Calcium 11.3 mg/dL (9.7-11.5 mg/dl)
the time of pacemaker placement. BUN 48 mg/dL (5-21 mg/dL)
Physical Examination: Creatinine 1.8 mg/dL (0.3-1.2 mg/dL)
Body Weight: 9.8 kg Glucose 81 mg/dL (64-123 mg/dL)
General: Quiet, alert, and responsive. Slightly tacky but pink Total protein 7.6 g/dL (5.4-7.6 g/dL)
mucous membranes, capillary refill time <2 s, approximately Albumin 2.7 g/dL (3.0-4.4 g/dL)
5% dehydrated, T = 104°F, HR = 160 beats/min, RR = 40 Globulin 4.9 g/dL (1.8-3.9 g/dL)
breaths/min. ALT 61 U/L (19-67 U/L)
Eyes, Ears, Nose, and Throat: A mild crusted ocular discharge AST 17 U/L (19-42 U/L)
was present bilaterally, as well as mild serous nasal discharge, ALP 190 U/L (21-170 U/L)
and moderate periodontal disease. Creatine kinase 61 U/L (51-399 U/L)
Integument: A 1-cm wound that discharged purulent material Gamma GT 3 U/L (0-6 U/L)
was present in the region of the right jugular incision, with a Cholesterol 323 mg/dL (135-361 mg/dL)
draining tract located along the jugular lead that extended Total bilirubin 0.1 mg/dL (0-0.2 mg/dL)
to the pacemaker generator over the right cervical region. Magnesium 1.7 mg/dL (1.5-2.6 mg/dL).
There was an accumulation of fluid that measured 1 cm in Urinalysis: SGr 1.013; pH 6.0, 150 mg/dL protein, no bilirubin,
diameter over the generator. 150 erythrocytes/µL hemoprotein, no glucose, no ketones,
Musculoskeletal: Ambulatory with normal gait, BCS 5/9 12-18 WBC/HPF, 8-12 RBC/HPF, no other significant findings.
Cardiovascular: Heart rate 160 beats/min, regular rhythm, Coagulation Panel: PT 8.7 s (7.0-9.3 s), APTT 14.1 s (10.4-12.9 s),
grade III/VI left apical systolic murmur, femoral pulses strong fibrinogen 412 mg/dL (109-311 mg/dL), D-dimers 418 ng/
and synchronous mL (0-186 ng/mL), antithrombin 84% (80%-120%).
Respiratory: Mild crackles were auscultated in the left and Imaging Findings:
right caudal lung fields. Thoracic Radiographs: See Figure 35-3. Mild cardiomegaly
Abdominal Palpation: Cranial organomegaly was present. was present. The far caudal left pulmonary vasculature
CHAPTER 35  Staphylococcus Infections 353

(≤0.5 µg/mL), and vancomycin (≤1.00 µg/mL). The Serratia


marcescens was resistant to cefazolin, cefoxitin, cephalothin,
and doxycycline; and susceptible to amikacin (≤4.00 µg/
mL), amoxicillin-clavulanic acid (4.00 µg/mL), ampicillin
(≤2.00 µg/mL), cefovecin (1.00 µg/mL), cefpodoxime (≤2.00
µg/mL), enrofloxacin (≤0.25 µg/mL), marbofloxacin (≤1
µg/mL), gentamicin (≤1 µg/mL), imipenem (≤1 µg/mL),
ticarcillin (≤8 µg/mL), ticarcillin-clavulanate (≤8 µg/mL), and
trimethoprim-sulfamethoxazole (≤0.5 µg/mL).
Bottle 2 (right lateral saphenous vein): Direct smear: small num-
bers of gram-positive cocci. Culture: coagulase-negative
Staphylococcus.
Urine aerobic culture and susceptibility (cystocentesis
specimen): >105 organisms/mL coagulase-negative
Staphylococcus, with an identical antibiogram to that of the
isolate from the blood.
Seroma Aerobic and Anaerobic Culture and Susceptibility:
Small numbers of coagulase-negative Staphylococcus, with
an identical antibiogram to that of the isolate from the
blood and urine.
Diagnosis: Bacteremia and probable embolic pneumonia
due to methicillin-resistant Staphylococcus spp. and possibly
Serratia marcescens associated with pacemaker placement.
Skin/soft tissue and urinary tract infection with methicillin-
resistant Staphylococcus spp.
Treatment: Elliot was hospitalized with full-contact
precautions that included the use of gloves and gowns and
contact with a limited number of individuals. Treatment was
initiated with intravenous fluids, vancomycin (10 mg/kg IV
FIGURE 35-3  Dorsoventral thoracic radiograph from a 10-year-old male neu- q6h) for the methicillin-resistant staphylococcal infection,
tered miniature schnauzer with methicillin-resistant Staphylococcus bacteremia and and enrofloxacin (10 mg/kg IV q24h) for a possible Serratia
pacemaker infection. There was a bronchointerstitial pattern and mild cardiomegaly. marcescens infection. After 48 hours of treatment, the
The caudal left pulmonary vasculature was indistinct with patchy infiltrates (arrows),
pacemaker was removed and replaced with a ventricular
which raised suspicion for septic embolic pneumonia.
(VVIR) epicardial pacemaker, and a Penrose drain was
placed at the incision site. A repeat blood culture (four
was indistinct with region of patchy infiltrates (arrows). bottles) performed 3 days later showed growth of the
The pacemaker was static in appearance with a generator CoNS in one bottle. The pulmonary infiltrates resolved,
embedded in the tissues dorsal to the cervical spine and two creatinine normalized, and repeat blood and urine cultures
leads extending into the region of the cardiac silhouette— were negative after 10 days of vancomycin treatment.
one terminating at the level of the right atrium and the The dog was discharged from the hospital. Follow-up
other terminating at the level of the right ventricle. Within cultures after an additional 10 days off antibiotics remained
the viewable abdomen, the liver was persistently markedly negative. Six months later, Elliot developed right-sided
enlarged and rounded. Given the absence of vasculature in congestive heart failure with ascites and pleural effusion.
the left caudal lung lobe, embolism in this area was suspected. Aerobic bacterial culture of the ascites fluid was negative.
Echocardiogram (to evaluate for endocarditis): Mild mitral On echocardiography, the tricuspid valve was severely
regurgitation was present, with no evidence of endocarditis. thickened and irregular. Endocarditis was suspected.
Microbiologic Testing: Additional diagnostics that included blood cultures were
Aerobic Blood Culture and Susceptibility: Bottle 1 (right declined by the owners, who elected euthanasia without
cephalic catheter): Direct smear from bottle: small numbers necropsy.
of gram-positive cocci and small numbers of gram-variable Comments: Vancomycin was used to treat this severe
rods. Culture: coagulase-negative Staphylococcus and methicillin-resistant staphylococcal infection because
Serratia marcescens. The CoNS was methicillin resistant the dog had renal failure and there was concern for
and resistant to clindamycin, enrofloxacin, marbofloxacin, aminoglycoside toxicity. The S. marcescens may have
erythromycin, trimethoprim-sulfamethoxazole; had represented a contaminant, but it was treated because of
intermediate susceptibility to gentamicin (8.00 µg/ the severity of the dog’s condition. Whether the endocarditis
mL); and was susceptible to amikacin (≤4.00 µg/mL), that developed was the result of a new infection or
chloramphenicol (8.00 µg/mL), doxycycline (4.00 µg/mL), persistence of the original staphylococcal infection could
rifampin (≤1 µg/mL), daptomycin (≤0.25 µg/mL), linezolid not be determined.
354 SECTION 2  Bacterial Diseases

SUGGESTED READING 15. Soares Magalhaes RJ, Loeffler A, Lindsay J, et al. Risk factors for
methicillin-resistant Staphylococcus aureus (MRSA) infection in
Papich MG. Selection of antibiotics for meticillin-resistant Staphylococ- dogs and cats: a case-control study. Vet Res. 2010;41:55.
cus pseudintermedius: time to revisit some old drugs? Vet Dermatol. 16. Girard C, Higgins R. Staphylococcus intermedius cellulitis and
2012;23(4):352-360. toxic shock in a dog. Can Vet J. 1999;40:501-502.
17. Weese JS, Poma R, James F, et al. Staphylococcus pseudintermedius
REFERENCES necrotizing fasciitis in a dog. Can Vet J. 2009;50:655-656.
1. Ogsten A. Micrococcus poisoning. J Anat Physiol. 1882;16:526. 18. Osland AM, Vestby LK, Fanuelsen H, et  al. Clonal diversity
2. Ghebremedhin B, Layer F, Konig W, et  al. Genetic classification and biofilm-forming ability of methicillin-resistant Staphylococ-
and distinguishing of Staphylococcus species based on different cus pseudintermedius. J Antimicrob Chemother. 2012 Apr, 67:
partial gap, 16S rRNA, hsp60, rpoB, sodA, and tuf gene sequences. 841-848.
J Clin Microbiol. 2008;46:1019-1025. 19. Lewis 2nd JS, Jorgensen JH. Inducible clindamycin resistance in
3. Cain CL, Morris DO, O’Shea K, et  al. Genotypic relatedness and staphylococci: should clinicians and microbiologists be concerned?
phenotypic characterization of Staphylococcus schleiferi subspecies in Clin Infect Dis. 2005;40:280-285.
clinical samples from dogs. Am J Vet Res. 2011;72:96-102. 20. Rubin JE, Ball KR, Chirino-Trejo M. Antimicrobial susceptibility
4. Litster A, Moss SM, Honnery M, et al. Prevalence of bacterial spe- of Staphylococcus aureus and Staphylococcus pseudintermedius
cies in cats with clinical signs of lower urinary tract disease: rec- isolated from various animals. Can Vet J. 2011;52:153-157.
ognition of Staphylococcus felis as a possible feline urinary tract 21. Faires MC, Gard S, Aucoin D, et al. Inducible clindamycin-resistance
pathogen. Vet Microbiol. 2007;121:182-188. in methicillin-resistant Staphylococcus aureus and methicillin-
5. Eckholm NG, Outerbridge CA, White SD, et al. Prevalence of and resistant Staphylococcus pseudintermedius isolates from dogs and
risk factors for infection with methicillin-resistant Staphylococcus cats. Vet Microbiol. 2009;139:419-420.
spp. in dogs with pyoderma in northern California. Submitted. 22. Lafaurie M, Porcher R, Donay JL, et  al. Reduction of fluoroqui-
2012. nolone use is associated with a decrease in methicillin-resistant
6. Devriese LA, Vancanneyt M, Baele M, et al. Staphylococcus pseud- Staphylococcus aureus and fluoroquinolone-resistant Pseudomo-
intermedius sp. nov., a coagulase-positive species from animals. Int nas aeruginosa isolation rates: a 10 year study. J Antimicrob Che-
J Syst Evol Microbiol. 2005;55:1569-1573. mother. 2012;67(4):1010-1015.
7. Devriese LA, Hermans K, Baele M, et  al. Staphylococcus pseud- 23. Falagas ME, Bliziotis IA, Fragoulis KN. Oral rifampin for eradica-
intermedius versus Staphylococcus intermedius. Vet Microbiol. tion of Staphylococcus aureus carriage from healthy and sick popu-
2009;133:206-207. lations: a systematic review of the evidence from comparative trials.
8. Ben Zakour NL, Bannoehr J, van den Broek AH, et al. Complete Am J Infect Control. 2007;35:106-114.
genome sequence of the canine pathogen Staphylococcus pseudin- 24. Jordan D, Simon J, Fury S, et al. Carriage of methicillin-resistant
termedius. J Bacteriol. 2011;193:2363-2364. Staphylococcus aureus by veterinarians in Australia. Aust Vet J.
9. Perreten V, Kadlec K, Schwarz S, et al. Clonal spread of methicillin- 2011;89:152-159.
resistant Staphylococcus pseudintermedius in Europe and North 25. Loeffler A, Pfeiffer DU, Lloyd DH, et al. Meticillin-resistant Staph-
America: an international multicentre study. J Antimicrob Che- ylococcus aureus carriage in UK veterinary staff and owners of
mother. 2010;65:1145-1154. infected pets: new risk groups. J Hosp Infect. 2010;74:282-288.
10. van Duijkeren E, Kamphuis M, van der Mije IC, et al. Transmis- 26. Burstiner LC, Faires M, Weese JS. Methicillin-resistant Staphylo-
sion of methicillin-resistant Staphylococcus pseudintermedius coccus aureus colonization in personnel attending a veterinary sur-
between infected dogs and cats and contact pets, humans and the gery conference. Vet Surg. 2010;39:150-157.
environment in households and veterinary clinics. Vet Microbiol. 27. Hanselman BA, Kruth SA, Rousseau J, et al. Methicillin-resistant
2011;150:338-343. Staphylococcus aureus colonization in veterinary personnel. Emerg
11. Griffeth GC, Morris DO, Abraham JL, et al. Screening for skin car- Infect Dis. 2006;12:1933-1938.
riage of methicillin-resistant coagulase-positive staphylococci and 28. Paul NC, Moodley A, Ghibaudo G, et al. Carriage of methicillin-
Staphylococcus schleiferi in dogs with healthy and inflamed skin. resistant Staphylococcus pseudintermedius in small animal vet-
Vet Dermatol. 2008;19:142-149. erinarians: indirect evidence of zoonotic transmission. Zoonoses
12. Morris DO, Lautenbach E, Zaoutis T, et al. Potential for pet ani- Public Health. 2011;58:533-539.
mals to harbour methicillin-resistant Staphylococcus aureus when 29. Morris DO, Boston RC, O’Shea K, et  al. The prevalence of car-
residing with human MRSA patients. Zoonoses Public Health. riage of meticillin-resistant staphylococci by veterinary derma-
2012;59(4):286-293. tology practice staff and their respective pets. Vet Dermatol.
13. Loeffler A, Pfeiffer DU, Lindsay JA, et  al. Lack of transmission 2010;21(4):400-407.
of methicillin-resistant Staphylococcus aureus (MRSA) between
apparently healthy dogs in a rescue kennel. Vet Microbiol.
2010;141:178-181.
14. Abraham JL, Morris DO, Griffeth GC, et al. Surveillance of healthy
cats and cats with inflammatory skin disease for colonization of the skin
by methicillin-resistant coagulase-positive staphylococci and Staphy-
lococcus schleiferi ssp. schleiferi. Vet Dermatol. 2007;18:252-259.
CHAPTER 36

Gram-negative Bacterial Infections


Jane E. Sykes

LPS is a large molecule that varies in composition from one


Overview of Gram-negative Bacteria bacterial species and strain to another. It contributes to the struc-
First Described: Gram-negative bacteria were first described in tural integrity of gram-negative bacteria and is a potent virulence
Berlin by the Danish scientist Hans Christian Gram in 1884 factor. It consists of the lipid A backbone, a core ­oligosaccharide,
when he used Gram stain to visualize ­Klebsiella ­pneumoniae and the O antigen side chain. Lipid A is a phosphorylated disac-
in the lungs of people who died of pneumonia.1 charide to which long, hydrophobic fatty acid chains are attached,
Causes: Examples include Escherichia coli, Salmonella, Entero- which anchor the LPS into the outer membrane (­Figures 36-1
bacter, Citrobacter, Klebsiella, Campylobacter, Pseudomonas, and 36-2). The lipid A component is also known as endotoxin
and Acinetobacter. because it is the biologically active portion of the molecule, in that
it stimulates a potent host inflammatory response (see Chapter
Geographic Distribution: Worldwide
86). The core oligosaccharide connects lipid A to the O ­antigen
Mode of Transmission: Direct contact or contact with conta­m­ side chain. Its composition differs between bacterial species. The
inated fomites; the bacteria often invade opportunistically O antigen is a repeating polysaccharide that projects from the
Major Clinical Signs: Affected sites include UTIs, wound surface of the bacterial cell and varies in length from 1 to 60
infections, pneumonia, pyothorax, peritonitis, pyometra, repeats. It is the antigenic portion of the molecule and is respon-
ocular surface infections, otitis externa, pyoderma, bacte- sible for serogroup classification of gram-negative bacteria.
remia, and endocarditis, but virtually any body system can Gram-negative cocci, such as Moraxella or Neisseria, are not
be affected. widely recognized as significant pathogens of dogs and cats. How-
ever, they are commensals of the oral cavity of dogs and cats, can
Differential Diagnoses: Infections with gram-positive bac-
cause bite wound infections in humans,2 and ­Neisseria canis was
teria such as Staphylococcus spp. and Streptococcus spp.;
isolated in pure culture from a deep mandibular abscess from one
to a lesser extent infections with nonbacterial pathogens
dog.3 Gram-negative rods are classified into Enterobacteriaceae
such as fungi.
and non-Enterobacteriaceae species. Enterobacteriaceae that
Human Health Significance: Gram-negative bacteria shed cause disease in dogs and cats include E ­ scherichia coli, ­Proteus,
by dogs and cats have the potential to colonize humans, ­Salmonella, ­Enterobacter, ­Citrobacter, Serratia, and Klebsiella.
especially those with impaired host defenses. They may Non-Enterobacteriaceae species include the Pasteurellaceae
also be responsible for infections of dog or cat bites. (­Pasteurella multocida), as well as P ­ seudomonas aeruginosa and
Acinetobacter. Other gram-negative bacteria are coccobacilli
or spiral-shaped organisms and include B ­ artonella, ­Bordetella,
Etiology, Epidemiology, and Clinical Features ­Campylobacter, ­Francisella, Helicobacter, and Brucella.
Like gram-positive cocci, some gram-negative bacteria have
Gram-negative bacteria are widespread causes of opportunistic tremendous ability to form biofilms, or bacterial aggregates
infections in dogs and cats and primarily cause disease when that are embedded in a matrix of exopolysaccharide (bacterial
host defenses are impaired. They are important causes of noso- slime), which form on living or nonliving surfaces. These pro-
comial disease. In contrast to gram-positive bacteria, gram-­ tect the bacteria, and bacteria in biofilms can resist the effect of
negative bacteria have a complex outer membrane that contains antimicrobial drugs and disinfectants.
lipopolysaccharide (LPS), as well as structures known as porins
that regulate transport of molecules in and out of the cell, which Enterobacteriaceae
includes antimicrobial drugs. Between this outer membrane and Many of the Enterobacteriaceae contribute to the normal gastro-
the inner (cytoplasmic) membrane is a periplasmic space, which intestinal flora of animals. Organisms that belong to the Entero-
contains the structural polymer peptidoglycan (or murein) bacteriaceae possess a capsule (K antigen) and may be motile via
(­Figure 36-1). The peptidoglycan layer of gram-negative bacte- flagella (H antigen). Some highly encapsulated species, such as
ria is much thinner than that of gram-positive bacteria, approx- Klebsiella and Enterobacter, have a mucoid appearance when
imately one layer, or 7 to 8 nm thick (compared with many they grow in culture. The flagella project from the bacterial
layers, or 20 to 80 nm thick for gram-positive bacteria). The cell and contain the filament-forming protein flagellin, which
inner cytoplasmic membrane contains many different proteins (along with LPS) can stimulate the host inflammatory response.
that are lipoproteins anchored into the outer lipid bilayer leaflet, The Enterobacteriaceae also possess fimbriae or pili, which are
transmembrane proteins, or peripheral membrane proteins that thinner than flagella and play a role in adhesion and sometimes
lie adjacent to the bilayer leaflets. bacterial aggregation. Finally, they possess a variety of complex

355
356 SECTION 2  Bacterial Diseases

A B
FIGURE 36-1  Structure of the envelope of gram-positive (A) and gram-negative (B) bacteria. The thickness of the peptidoglycan layer is greater in gram-positive bacteria than in
gram-negative bacteria.

secretion systems (type I, II, III secretion systems). These facili- P fimbriae, hemolysin, and cytotoxic necrotizing factor. Specific
tate export of toxins such as hemolysins, and molecules that virulence factors, as well as biofilm-forming capacity, have been
are inserted into, and form pores in, host cell membranes. The associated with persistent or relapsing UTIs,12,13 and uropatho-
bacteria then use these pores to inject toxins directly into the genic E. coli can invade and persist within bladder epithelial
host cell. The Enterobacteriaceae also have a variety of iron cells. In one study, canine UTIs associated with E. coli were more
scavenging systems, which are important virulence factors, and likely to be recurrent than those associated with other uropatho-
transmissible plasmids that contain genes for virulence factors gens.14 There has been concern that dogs may act as a source
and antimicrobial resistance factors. of E. coli strains that are pathogenic for humans.11,15 In other
extraintestinal E. coli infections, E. coli more closely resemble
Escherichia coli commensal strains, and host factors may be more important
Escherichia coli is the most common cause of urinary tract than bacterial virulence factors. The reader is referred to Chapter
infections (UTIs) in dogs and cats and can also be isolated from 46 for more information on intestinal E. coli infections.
dogs and cats with pyometra, prostatitis, peritonitis, cholangi-
tis, cholecystitis, bacteremia and endocarditis, bronchopneumo- Proteus
nia (such as that secondary to aspiration), and pyothorax. There Proteus mirabilis is a common cause of upper and lower UTIs
is one report of necrotizing fasciitis in a dog associated with E. in dogs as well as otitis externa and uncommonly pyoderma.
coli infection.4 It produces a potent urease and so may contribute to devel-
Antibiotic resistance in E. coli isolates from dogs and cats opment of struvite urolithiasis. Uroliths harbor bacteria that
is an increasing problem; 51% of 376 isolates collected from can emerge as the uroliths dissolve. Proteus has a character-
sick dogs and cats in the United States in 2005 showed resis- istic swarming motility and may obscure the growth of other
tance to at least one drug, and 29% of the 376 isolates were co-infecting microorganisms when grown in the laboratory on
multiple drug resistant (MDR; defined as resistant to three or agar. ­Proteus penneri, an uncommon cause of nosocomial infec-
more classes of antimicrobial drugs).5 The prevalence of resis- tions in humans, has rarely been isolated from wound infections
tance in 395 isolates from healthy dogs and cats from Canada in dogs seen at the author’s hospital.
was lower, 19%.6 Extended-spectrum β-lactamase enzyme pro-
duction has also been identified in E. coli isolates from dogs Klebsiella
and cats in the United States,7 and a few isolates were found The most common species of Klebsiella that infects dogs and
to express New Dehli metallo-β-lactamase (NDM-1), which cats is K. pneumoniae. K. oxytoca is a nosocomial pathogen in
hydrolyses carbapenems.8 Nosocomial infections with MDR E. humans and has been reported in association with intravenous
coli have also been described in dogs and cats. catheter colonization in puppies with parvoviral enteritis.16
In humans, strains of E. coli that cause UTIs differ from those K. pneumoniae primarily causes upper and lower UTIs in
that cause diarrhea, and there is some evidence that this may also dogs and cats, but can also cause pneumonia, sometimes with
be true for strains that cause UTIs versus pyometra in dogs and abscessation (Figures 36-3 and 36-4). It has also been isolated
cats.9-11 Strains of E. coli that cause UTIs belong to the pathotype from dogs with hepatic abscesses.17 Nosocomial infections may
extraintestinal pathogenic E. coli (ExPEC). These strains possess be associated with bacteremia, endocarditis, and postoperative
a number of virulence factors, such as siderophores, type 1 and skin and soft tissue infections. The capsule of K. pneumoniae is
CHAPTER 36  Gram-negative Bacterial Infections 357

FIGURE 36-3  Lungs of an 11-year-old intact male German shepherd dog with
severe, acute, suppurative, necrotizing and hemorrhagic bronchopneumonia due to Kleb­
siella pneumoniae infection. The lung lobes are severely congested. Five days before death,
multiple laminectomies had been performed for intervertebral disc disease, and 2 days
later the dog developed aspiration pneumonia that necessitated mechanical ventilation.
Euthanasia was elected when there was further deterioration in the dog’s condition, and
bloody exudate was present in the endotracheal tube just before euthanasia.

FIGURE 36-2  Structure of bacterial lipopolysaccharide.

FIGURE 36-4  Histopathology of the lung from a 4-month-old intact male New-
a major virulence factor; it inhibits phagocytosis and is respon- foundland dog with severe, multifocal, necrotizing lung abscesses. Intralesional rod-shaped
sible for the mucoid appearance of the organism in culture ­bacteria were also present. Klebsiella pneumoniae was isolated from the lesions. H&E stain,
40× magnification.
(­Figure 36-5, A).
All strains of K. pneumoniae are resistant to ampicillin,
because they possess a chromosomal gene that encodes a penicillin often linked to intravenous drug use. The organism has a tremen-
β-lactamase. Some isolates, such as nosocomial isolates or those dous ability to survive in the environment and may contaminate
from persistent UTIs, possess plasmids that contain resistance and remain viable in disinfectant solutions.18 It sometimes grows as
genes for multiple antimicrobial drugs. Of greatest concern are a contaminant, such as in blood cultures. Some, but not all, strains
Klebsiella isolates that express extended-spectrum β-lactamase of S. marcescens produce a red pigment when grown in culture.
(ESBL) enzymes, which hydrolyze third-generation cephalospo- In dogs, S. marcescens infection has been associated with
rins such as cefotaxime or ceftriaxone. The genes that encode UTIs, bacteremia and aortic valve endocarditis,19 catheter-
ESBLs are often located on plasmids that also contain genes for related infections in canine parvoviral enteritis,14 and necro-
aminoglycoside resistance. Although MDR K. pneumoniae are tizing fasciitis.20 Contamination of blood products was linked
not uncommonly isolated from dogs and cats, ESBL-producing to an outbreak of bacteremia in cats.21 Serratia is resistant to
K. pneumoniae isolates are rarely encountered. Treatment options ampicillin and first-generation cephalosporins as a result of a
for MDR K. pneumoniae may be limited to aminoglycosides, car- chromosomal β-lactamase. It may also acquire plasmid-mediated
bapenems, and sometimes trimethoprim-sulfamethoxazole. resistance to other antimicrobial drugs.

Serratia marcescens Enterobacter


Serratia marcescens is an important cause of nosocomial infections Enterobacter cloacae and, to a lesser extent, Enterobacter aero-
in both human and veterinary medicine. In human patients it is genes represent the majority of Enterobacter species isolated
358 SECTION 2  Bacterial Diseases

A B C
FIGURE 36-5  Gram-negative aerobic bacteria growing on blood agar. A, Mucoid colonies of Klebsiella pneumoniae. B, Pseudomonas aeruginosa. Transillumination using a light box
revealed the presence of hemolysis and production of a green pigment (pyoverdin). C, The plate in (B) had a fruity odor and a “mother-of-pearl” appearance when not transilluminated.

from dogs and cats in the author’s hospital. Enterobacter spp.


are mainly isolated from dogs and cats with UTIs but occasion-
ally cause wound or catheter-related infections. They generally
are intrinsically resistant to ampicillin and first-generation ceph-
alosporins and may also be resistant to multiple other antimi-
crobial drug classes.

Citrobacter
Citrobacter species are named for their ability to use citrate as a
sole carbon source. Citrobacter freundii is most commonly iso-
lated from dogs and cats with UTIs but can also cause catheter-
related infections, bacteremia, and endocarditis.22,23 ­Citrobacter
koseri was isolated from puppies with myocarditis.24 Like F
­Enterobacter and Serratia, Citrobacter are usually resistant to A
ampicillin and first-generation cephalosporins and may acquire
a number of plasmid-mediated resistance genes, including those
that encode ESBLs and resistance to fluoroquinolones.

Non-Enterobacteriaceae
Pasteurella
Bacteria that belong to the family Pasteurellaceae are small, fas-
tidious, nonmotile bacilli or coccobacilli that are commensals of
the oral cavities and upper respiratory tracts of dogs and espe-
cially cats. The most common species of Pasteurella associated P
with disease in dogs and especially cats is Pasteurella multocida.
Other species include Pasteurella canis, Pasteurella dagmatis,
and Pasteurella stomatis. Pasteurella spp. have been associated
with a wide variety of disease manifestations that include pyo- B
thorax, upper respiratory tract infections (usually secondary to
viral infections or chronic rhinosinusitis), bronchopneumonia, FIGURE 36-6  Electron micrograph of Pseudomonas aeruginosa showing ultrastructural
UTIs, ocular surface infections, wound infections, cutaneous features. A, Single cell with polar flagellum (F) with part of the flagellum running under the
abscesses, otitis externa, bacteremia, and, rarely, infective endo- cell body. B, Two cells showing thin, hair-like pili (P). (A, Courtesy of Dr. Steve Lory. In Mandell
GL, Bennett JE, Dolin R, eds. Mandell, Douglas and Bennett’s principles and practice of infec-
carditis. They may be found in mixed infections with other bac-
tious diseases, 7 ed. Philadelphia; Churchill Livingstone Elsevier; 2010:2835-2860; B, Courtesy
teria, such as Streptococcus canis, other gram-negative bacteria, of Dr. Martin Lee and Dr. Milan Bajmoczi, Harvard Medical School. In: Longo D, Fauci A, Kasper
or anaerobes. In humans, they are an important cause of animal D, et al. Harrison’s principles of internal medicine, 18 ed. Vol 1, New York; McGraw-Hill; 2012.)
bite wound infections.2 Pasteurella spp. are usually broadly sus-
ceptible to antimicrobial drugs, and the treatment of choice is a
penicillin such as penicillin G, ampicillin, or amoxicillin.
(blue), pyorubin (red), pyomelanin (black), and pyoverdin (yel-
Pseudomonas aeruginosa low-green to yellow-brown and fluorescent), and for its charac-
Pseudomonas aeruginosa is the main pathogenic species in the teristic “corn taco” or “grape”-like odor (see Figure 36-5, B and
family Pseudomonaceae. It is known for its ability to produce a C). P. aeruginosa has one polar flagellum and is covered in thin,
variety of pigments when grown in culture, such as pyocyanin hairlike pili (Figure 36-6). It can also produce an extracellular
CHAPTER 36  Gram-negative Bacterial Infections 359

polysaccharide known as alginate, which results in a mucoid Investigation of suspected nosocomial outbreaks of
colony appearance. P. aeruginosa infection generally requires typing of isolates
Pseudomonas aeruginosa is widespread in the environment from affected animals and the environment with molecular
and can grow in a huge variety of different conditions, reflect- methods such as pulsed-field gel electrophoresis or multi-
ing its importance as a nosocomial pathogen. It is commonly locus sequence typing. Use of antibiograms alone does not
found on plants, water, and soil. It can survive in disinfectants adequately discriminate between strains.
and grow in diesel and jet fuel. It is occasionally isolated from
the skin, mucous membranes, and feces of healthy animals and Acinetobacter
humans. Almost any site of the body that is subject to injury Like P. aeruginosa, Acinetobacter is an important nosoco-
can become colonized and subsequently infected. In humans, mial opportunistic pathogen. Acinetobacter is present in soil
community-acquired infections can occur as a result of expo- and water and can be found in frozen foods, pasteurized milk,
sure to contaminated hot tubs, swimming pools, and contact and an enormous array of inert objects in the hospital environ-
lens solutions. In the hospital environment, P. aeruginosa ment, where it may persist for months. It colonizes the skin and
often colonizes moist environments such as sinks, endotracheal mucous membranes of healthy humans as well as those of dogs
tubes, and ventilator equipment. It also colonizes damp, mac- and cats.26,27 The main species that cause disease in animals are
erated tissues such as the ear canals, damaged cornea, trau- Acinetobacter baumannii and, to a lesser extent, ­Acinetobacter
matized skin (such as wounds and burns), and the interdigital lwoffii and Acinetobacter calcoaceticus. Multi­drug resistant
space. Because the organism can be resistant to a large number isolates of A. baumannii have been responsible for outbreaks
of different antimicrobial drugs, repeated treatment of second- of nosocomial infections in veterinary hospitals28,29 and, like
ary bacterial infections in sites where impaired host defenses MDR P. aeruginosa, may be resistant to a huge variety of anti-
exist with a variety of antimicrobials can select for MDR P. microbial drug classes. Carbapenem-resistant A. ­baumannii
aeruginosa infection at these sites. Examples of such conditions have been isolated from humans as well as animals.29,30 Clini-
in dogs and cats include recurrent aspiration pneumonia and/ cal manifestations of Acinetobacter infections in dogs and
or bronchiectasis, cystitis, chronic feline rhinosinusitis, and oti- cats include nosocomial UTIs (e.g., after invasive urinary pro-
tis externa/media. cedures such as catheterization), surgical wound infections,
Pseudomonas aeruginosa possesses virtually every type pneumonia, catheter-related bacteremia, and, rarely, endocar-
of bacterial virulence factor, including exotoxins, a type III ditis. Necrotizing fasciitis associated with A. baumannii infec-
secretion system, lipopolysaccharide, pili, flagella, several tion was described in a cat.31 When Acinetobacter is grown
proteases, phospholipases, iron-scavenging mechanisms such from a specimen, care should be taken to differentiate infection
as pyoverdin production, and the ability to form extensive from contamination. Most veterinary isolates are susceptible
biofilms. Alginate protects the organism from phagocytosis, to aminoglycosides and carbapenems; some are susceptible to
and the pigment pyocyanin produces reactive oxygen species, ­trimethoprim-sulfamethoxazole and fluoroquinolones.28
which in turn damage host cells. Multidrug resistance results
from the production of specific β-lactamase enzymes (which Diagnosis
can include ESBLs), antibiotic efflux pumps, enzymes that
modify aminoglycosides or alter antibiotic binding sites (such Laboratory Abnormalities
as DNA gyrase for fluoroquinolones), and decreased bacterial Complete Blood Count, Biochemistry, and Urinalysis Findings
permeability. Dogs and cats with superficial gram-negative infections of sites
Infections with broadly susceptible P. aeruginosa isolates that drain to the exterior, such as wounds, the external ear canal,
could be treated with an antipseudomonal β-lactam (e.g., ticar- the nasal cavity, and the lower urinary tract, often have no clini-
cillin, a carbapenem, cefepime, ceftazidime) alone or in com- cally significant laboratory abnormalities. Deep tissue or sys-
bination with an aminoglycoside or a fluoroquinolone. The temic infections with gram-negative bacteria may be associated
combination of an aminoglycoside and an antipseudomonal with marked neutrophilia and a left shift, toxic neutrophils, or
penicillin has synergistic activity against some P. aeruginosa a degenerative left shift, sometimes with circulating metamyelo-
infections. Currently, most MDR P. aeruginosa isolates from cytes. Thrombocytopenia may be present in dogs and cats that
dogs and cats remain susceptible to carbapenems (such as develop disseminated intravascular coagulation (DIC). Anemia
meropenem) and aminoglycosides (especially amikacin), and of inflammatory disease, lymphopenia, and monocytosis may
so the usual treatment of choice is 1) a combination of these be present. The urinalysis of dogs and cats with gram-­negative
two drugs, 2) removal of any foreign or necrotic material and, bacterial UTIs may reveal pyuria, proteinuria, hematuria, and
3) if possible, resolution or management of any other underlying sometimes bacterial rods on sediment examination.
cause for impaired host defenses. Because parenterally admin-
istered aminoglycosides do not effectively penetrate airway Coagulation Profile
mucus, nebulized tobramycin has been used to treat resistant Systemic gram-negative bacterial infections may be associated
P. aeruginosa infections of the airways in human patients, but with evidence of DIC due to activation of the coagulation cas-
these may not adequately penetrate consolidated lung tissue.25 cade by endotoxin.
Effective treatment of MDR P. aeruginosa infections when
they occur in locations such as the feline nasal cavity may be Cytologic Abnormalities
extremely difficult, if not impossible, because of irreversible tur- Aspirates of affected tissues or pus, or transtracheal wash or
binate loss, the requirement for injectable antimicrobial drugs, bronchoalveolar lavage fluid specimens from dogs and cats with
poor penetration of aminoglycosides to the site of infection, and gram-negative bacterial infections may contain large numbers
an inadequate ability to effectively treat all surfaces of the nasal of degenerate and nondegenerate neutrophils, sometimes with
cavity with topical aminoglycosides. bacterial rods.
360 SECTION 2  Bacterial Diseases

TABLE 36-1
Examples of Systemic Antimicrobial Drugs That Could Be Selected for Treatment of Gram-Negative Bacterial Infections
in Dogs and Cats (See Also Chapter 8)
Infecting Species Drug Dose (mg/kg) Interval (hours) Route
Pasteurella Amoxicillin 20 8 PO
Ampicillin 20 6-8 IV, IM, SC
Penicillin G 20,000-40,000 U/kg 6-8 IV, IM
Susceptible Trimethoprim-­sulfamethoxazole 30 12 PO, IV
­Enterobacteriaceae Amoxicillin–clavulanate 20 8 PO
­potassium
Ciprofloxacin 20-30 24 PO
10 24 IV
MDR ­Enterobacteriaceae, Amikacin* 15-30 (dogs), 10-14 (cats) 24 IV, IM, SC
Pseudomonas, or Gentamicin* 9-14 (dogs), 5-8 (cats) 24 IV, IM, SC
­Acinetobacter Ticarcillin disodium, ticarcillin- 33-50 4-6 IV
clavulanate
Meropenem 12 8 SC
25 8 IV
Ceftazidime 30 6 IV

*Use aminoglycosides in combination with an antipseudomonal β-lactam such as ticarcillin, meropenem, or a third-generation cephalosporin.

Microbiologic Tests amoxicillin, or clavulanic acid–amoxicillin are reasonable initial


Isolation and Identification choices for treatment of UTIs associated with bacterial rods.32
Gram-negative bacteria are readily isolated from aspirates, tis- Treatment should also be based on knowledge of local resistance
sue biopsies, body fluid, blood, and lavage specimens. MacCon­ patterns among gram-negative bacteria, which may vary from
key and blood agar are usually used to isolate gram-negative region to region and practice to practice.
bacteria, which usually grow within 24 to 48 hours and are sub-
sequently differentiated on the basis of colony morphology and Prevention
biochemical reactions. Pasteurella spp. are more fastidious, and
species that infect dogs and cats do not grow on MacConkey Prevention of gram-negative infections involves resolution of
agar. Evaluation of antimicrobial susceptibility is important for impairment in host defenses and strict hospital infection control
all Enterobacteriaceae, P. aeruginosa, and Acinetobacter spp., practices (see Chapter 11).
given their propensity to develop resistance to many antimicro-
bial drug classes. Public Health Aspects
Treatment and Prognosis Strains of E. coli and other MDR gram-negative bacteria
harbored by dogs resemble pathogenic strains isolated from
The approach to treatment of gram-negative infections depends humans.15,30 As a result, concern has been raised that compan-
on the site of infection and the severity of disease (Table 36-1); the ion animals may be a source of organisms for human infection,
reader is referred to Chapters 84 to 90 for treatment recommenda- especially those that reside with humans with compromised
tions for different organ systems. Identification and management host defenses.
of the underlying cause are also important to minimize selection Pasteurella multocida is the most commonly isolated organ-
for resistant organisms. Any foreign material (such as catheters, ism from human dog and cat bite wounds.2 Pasteurella spp. can
implants) must be removed, and necrotic tissue debrided. When also colonize the respiratory tracts of humans with underlying
systemic antimicrobial drug treatment is required, it should pref- respiratory disease such as bronchiectasis or chronic sinusitis,
erably be based on the results of susceptibility testing, although and they occasionally have been reported in association with
Pasteurella spp. are generally susceptible to penicillin or amino­ peritonitis, bacteremia, and endocarditis. Most of these humans
penicillins such as amoxicillin. Because gram-negative bacte- have had contact with domestic animals in the household, and
ria have higher minimum inhibitory concentrations (MICs) to isolates from affected humans have been shown by molecu-
penicillins than do gram-positive bacteria, the high end of the lar methods to be identical to those colonizing animals in the
dose range and more frequent administration may be necessary. household.33
Some Enterobacteriaceae are intrinsically resistant to ampicillin Thus, immunocompromised owners or those with impaired
and first-generation cephalosporins, and initial treatment of seri- anatomic barriers should be instructed to wash their hands after
ous infections while awaiting the results of susceptibility testing handling their pet and perform regular household cleaning and
should include an aminoglycoside, a third-generation cephalo- disinfection. Companion animals should not be allowed to lick
sporin, or a fluoroquinolone. Trimethoprim-sulfamethoxazole, open wounds or the owner’s face.
CHAPTER 36  Gram-negative Bacterial Infections 361

CASE EXAMPLE Phosphorus 5.8 mg/dL (3.2-6.3 mg/dL)


Calcium 10.2 mg/dL (9.0-10.9 mg/dl)
Signalment: “Lottie”, a middle-aged, female spayed domestic BUN 49 mg/dL (18-33 mg/dL)
shorthair from northern California Creatinine 1.4 mg/dL (1.1-2.2 mg/dL)
History: Lottie was evaluated for chronic nasal discharge. Since Glucose 128 mg/dL (63-118 mg/dL)
her rescue as a stray 2.5 years earlier, intermittent sneezing Total protein 8.7 g/dL (6.6-8.4 g/dL)
and bilateral mucopurulent nasal discharge had been noted, Albumin 3.6 g/dL (2.2-4.6 g/dL)
and the discharge sometimes contained blood. Ten months Globulin 5.1 g/dL (2.8-5.4 g/dL)
before the evaluation, Lottie had been anesthetized for a nasal ALT 33 U/L (27-101 U/L)
flush at a local veterinary clinic, but developed respiratory AST 21 U/L (17-58 U/L)
distress on recovery and was hospitalized with oxygen ALP 36 U/L (14-71 U/L)
supplementation for 24 hours. She was ataxic, leaned to one Gamma GT 0 U/L (0-4 U/L)
side, and did not eat well for days, but over the subsequent Cholesterol 192 mg/dL (89-258 mg/dL)
months the ataxia and leaning gradually resolved. For the two Total bilirubin 0.1 mg/dL (0-0.2 mg/dL).
weeks before the evaluation Lottie had stertorous respiration Urinalysis: USG 1.044, pH 6, 75 mg/dL protein, no glucose,
and intermittent episodes of open-mouth breathing. For the bilirubin or ketones, 250 erythrocytes/µL hemoprotein,
past 9 months, Lottie had been treated with marbofloxacin >100 RBC/HPF (traumatic cystocentesis), 0-1 WBC/HPF.
and prednisolone. Before that, treatments for the nasal Imaging Findings:
discharge included clavulanic acid–amoxicillin, doxycycline, Thoracic Radiographs: Cardiovascular structures were within
or azithromycin, as well as l-lysine supplementation. Lottie normal limits. There was a small amount of air in the cervical
lived with six other cats, all of which had access to the esophagus (likely due to aerophagia) and a mild diffuse
outdoors, and she was fed commercial canned and dry cat bronchointerstitial pattern that was attributed to an obese
food. The FeLV and FIV status of these cats was unknown. body condition.
Current medications: marbofloxacin (5 mg/kg PO q24h) and Skull Computed Tomography Scan: Contiguous transverse
prednisolone (1 mg/kg PO q24h) 0.6-mm collimated images of the skull were available for
Physical Examination: review without contrast administration. There was moderate
Body Weight: 4 kg turbinate destruction of both right and left nasal passages
General: Bright, alert and responsive, hydrated. T = 100.3°F with lobular soft tissue attenuating material present along
(37.9°C), P = 220 beats/min, purring, body condition score 7/9. their length (­Figure 36-7, A). There was stenosis of both
Eyes, Ears, Nose, and Throat: Mucopurulent nasal discharge frontal sinuses to the point where the left frontal sinus was
was present that was most profuse on the left side. Nasal completely occluded. The right frontal sinus was almost
airflow was absent on the left side. Mild dental calculus was completely filled with soft tissue attenuating material.
present, but there were no oral masses or ulceration. The ear canals were within normal limits bilaterally. The
Respiratory: An inspiratory stertor was present, and referred mandibular lymph nodes were prominent but symmetrical.
upper airway noises were detected on auscultation of the Rhinoscopy: Severe turbinate destruction with diffuse
thorax. hyperemia and large accumulations of inspissated green
Cardiovascular, Gastrointestinal and Genitourinary, and mucus was present throughout the nasal passages (­Figure
Lymph Nodes: No clinically significant abnormalities were 36-7, B). Several biopsies of the right and left nasal cavities
detected. It was difficult to palpate abdominal structures were obtained for histopathology.
due to the presence of abdominal fat. Microbiologic Testing: Aerobic bacterial culture and
Neurologic Examination: There was a mild, intermittent, left- Mycoplasma culture (nasal brush specimen): a direct smear
sided head tilt. No other abnormalities were detected on full showed moderate numbers of neutrophils, but no organisms
neurologic examination. were seen. Moderate numbers of Pseudomonas aeruginosa
Laboratory Findings: were isolated. The P. aeruginosa was resistant to amoxicillin–
CBC: clavulanic acid, ampicillin, cefazolin, cefovecin, cefoxitin,
HCT 39.1% (30%-50%) cefpodoxime, ceftiofur, chloramphenicol, doxycycline,
MCV 44.7 fL (42-53 fL) enrofloxacin, marbofloxacin, ticarcillin, ticarcillin–clavulanic
MCHC 29.9 g/dL (30-33.5 g/dL) acid, and trimethoprim-sulfamethoxazole. It was susceptible
Reticulocytes 111,400 cells/µL (7000-60,000 cells/µL) only to amikacin (≤4 µg/mL), gentamicin (2 µg/mL), and
WBC 21,210 cells/µL (4500-14,000 cells/µL) imipenem (≤1 µg/mL).
Neutrophils 16,713 cells/µL (2000-9000 cells/µL) Histopathology of Nasal Biopsy Specimens: The
Lymphocytes 4115 cells/µL (1000-7000 cells/µL) submucosa was markedly expanded and multifocally
Monocytes 297 cells/µL (50-600 cells/µL) effaced by sheets of lymphocytes and plasma cells,
Eosinophils 64 cells/µL (150-1100 cells/µL) with neutrophils and fewer numbers of mast cells
Basophils 0 cells/µL (0-50 cells/µL) surrounding irregular spicules of woven bone extending
Platelets 316,000/µL (180,000-500,000 platelets/µL). from primary turbinates. Less affected sections contain
Serum Chemistry Profile: similar, predominately lymphoplasmacytic infiltrates
Sodium 151 mmol/L (151-158 mmol/L) that separated submucosal glands and elevated the
Potassium 3.7 mmol/L (3.6-4.9 mmol/L) superficial epithelium. Pathologic diagnosis: severe, chronic,
Chloride 113 mmol/L (117-126 mmol/L) and extensive lymphoplasmacytic and neutrophilic
Bicarbonate 19 mmol/L (15-21 mmol/L) rhinitis with turbinate remodeling.

Continued
362 SECTION 2  Bacterial Diseases

L R

A A B
FIGURE 36-7  Nasal cavity of a middle-aged, female spayed domestic shorthair with chronic rhinosinusitis and secondary infection with multidrug-resistant Pseudomonas aeru­
ginosa. A, Computed tomographic scan showing destruction of the turbinates of the left and right nasal passages. B, Rhinoscopic findings included severe turbinate destruction,
hyperemia, and accumulations of thick green mucus.

Diagnosis: Feline chronic rhinosinusitis with suspected to play a role. Severe turbinate destruction predisposes to
opportunistic MDR P. aeruginosa infection. secondary opportunistic infections with a variety of bacteria,
Treatment and Outcome: The nasal cavity was lavaged. The which include staphylococci, Bordetella bronchiseptica,
prednisolone was tapered over 1 week, then discontinued, Streptococcus canis, Mycoplasma spp., E. coli, Pasteurella
and 7 days later, treatment with piroxicam was commenced multocida, and P. aeruginosa. In this cat, selection for MDR
(0.3 mg/kg PO q24h for 7 days, then q48h thereafter). Other P. aeruginosa likely resulted from repeated and chronic use
medications discussed with the owner were N-acetylcysteine of a variety of antimicrobial drugs. The reticulocytosis and
and intranasal gentamicin drops, but the prognosis given for increased serum BUN concentration likely was secondary to
disease resolution was poor. chronic epistaxis and swallowed blood. The cat’s neurologic
Comments: The underlying cause of feline chronic rhinosinusitis signs and head tilt were thought to be due to peripheral
is poorly defined, but infection of kittens with respiratory vestibular disease, possibly due to otitis media, but there was
viruses such as feline herpesvirus 1 (FHV-1) is suspected no evidence of middle ear disease on the CT scan.

SUGGESTED READINGS
6. Leonard EK, Pearl DL, Finley RL, et  al. Comparison of antimi-
Francey T, Gaschen F, Nicolet J, et al. The role of Acinetobacter bau- crobial resistance patterns of Salmonella spp. and Escherichia coli
mannii as a nosocomial pathogen for dogs and cats in an intensive recovered from pet dogs from volunteer households in Ontario
care unit. J Vet Intern Med. 2000;14:177-183. (2005-06). J Antimicrob Chemother. 2012;67:174-181.
Hohenhaus AE, Drusin LM, Garvey MS. Serratia marcescens contami- 7. Shaheen BW, Nayak R, Foley SL, et  al. Molecular characteriza-
nation of feline whole blood in a hospital blood bank. J Am Vet Med tion of resistance to extended-spectrum cephalosporins in clinical
Assoc. 1997;210:794-798. ­Escherichia coli isolates from companion animals in the United
States. Antimicrob Agents Chemother. 2011;55:5666-5675.
REFERENCES 8. Shaheen BW, Nayak R, Boothe DM. First reported case of New
1. Gram C. Über die isolierte Färbung der Schizomyceten in Schnitt- und Delhi Metallo (NDM) carpabenem-positive gene in Escherichia
Trockenpräparaten [in German]. Fortschr Med. 1884;2:185-189. coli from companion animals in the United States. San Francisco,
2. Talan DA, Citron DM, Abrahamian FM, et al. Bacteriologic analy- USA: Proceedings of the 52nd ICAAC symposium; 2012Abstract
sis of infected dog and cat bites. Emergency Medicine Animal Bite #157.
Infection Study Group. N Engl J Med. 1999;340:85-92. 9. Chen YM, Wright PJ, Lee CS, et al. Uropathogenic virulence ­factors
3. Cantas H, Pekarkova M, Kippenes HS, et al. First reported isola- in isolates of Escherichia coli from clinical cases of canine pyometra
tion of Neisseria canis from a deep facial wound infection in a dog. and feces of healthy bitches. Vet Microbiol. 2003;94:57-69.
J Clin Microbiol. 2011;49:2043-2046. 10. Freitag T, Squires RA, Schmid J, et al. Feline uropathogenic Esch-
4. Worth AJ, Marshall N, Thompson KG. Necrotising fasciitis associ- erichia coli from Great Britain and New Zealand have dissimilar
ated with Escherichia coli in a dog. N Z Vet J. 2005;53:257-260. virulence factor genotypes. Vet Microbiol. 2005;106:79-86.
5. Shaheen BW, Boothe DM, Oyarzabal OA, et  al. Antimicrobial 11. Siqueira AK, Ribeiro MG, Leite Dda S, et  al. Virulence factors
resistance profiles and clonal relatedness of canine and feline in Escherichia coli strains isolated from urinary tract infection
­Escherichia coli pathogens expressing multidrug resistance in the and pyometra cases and from feces of healthy dogs. Res Vet Sci.
United States. J Vet Intern Med. 2010;24:323-330. 2009;86:206-210.
CHAPTER 36  Gram-negative Bacterial Infections 363

12. Ejrnaes K, Stegger M, Reisner A, et  al. Characteristics of Esche- 24. Cassidy JP, Callanan JJ, McCarthy G, et al. Myocarditis in sibling
richia coli causing persistence or relapse of urinary tract infections: boxer puppies associated with Citrobacter koseri infection. Vet
phylogenetic groups, virulence factors and biofilm formation. Viru- Pathol. 2002;39:393-395.
lence. 2011;2:528-537. 25. Pier GB, Ramphal R. Pseudomonas aeruginosa. In: Mandell GL,
13. Soto SM, Smithson A, Horcajada JP, et  al. Implication of bio- Bennett JE, Dolin R, eds. Mandell, Douglas and Bennett’s Prin-
film formation in the persistence of urinary tract infection ciples and Practice of Infectious Diseases. 7th ed. Philadelphia, PA:
caused by uropathogenic Escherichia coli. Clin Microbiol Infect. Elsevier; 2010:2835-2860.
2006;12:1034-1036. 26. Saphir DA, Carter GR. Gingival flora of the dog with special
14. Ball KR, Rubin JE, Chirino-Trejo M, et al. Antimicrobial resistance reference to bacteria associated with bites. J Clin Microbiol.
and prevalence of canine uropathogens at the Western College of 1976;3:344-349.
Veterinary Medicine Veterinary Teaching Hospital, 2002-2007. 27. Krogh HV, Kristensen S. A study of skin diseases in dogs and cats.
Can Vet J. 2008;49:985-990. II. Microflora of the normal skin of dogs and cats. Nord Vet Med.
15. Harada K, Okada E, Shimizu T, et  al. Antimicrobial resistance, 1976;28:459-463.
virulence profiles, and phylogenetic groups of fecal Escherichia coli 28. Zordan S, Prenger-Berninghoff E, Weiss R, et  al. Multidrug-
isolates: a comparative analysis between dogs and their owners in resistant Acinetobacter baumannii in veterinary clinics. Germany.
Japan. Comp Immunol Microbiol Infect Dis. 2012;35:139-144. Emerg Infect Dis. 2011;17:1751-1754.
16. Lobetti RG, Joubert KE, Picard J, et  al. Bacterial colonization of 29. Francey T, Gaschen F, Nicolet J, et al. The role of Acinetobacter
intravenous catheters in young dogs suspected to have parvoviral baumannii as a nosocomial pathogen for dogs and cats in an inten-
enteritis. J Am Vet Med Assoc. 2002;220:1321-1324. sive care unit. J Vet Intern Med. 2000;14:177-183.
17. Farrar ET, Washabau RJ, Saunders HM. Hepatic abscesses in dogs: 30. Endimiani A, Hujer KM, Hujer AM, et  al. Acinetobacter bau-
14 cases (1982-1994). J Am Vet Med Assoc. 1996;208:243-247. mannii isolates from pets and horses in Switzerland: molecu-
18. Fox JG, Beaucage CM, Folta CA, et al. Nosocomial transmission of lar characterization and clinical data. J Antimicrob Chemother.
Serratia marcescens in a veterinary hospital due to contamination 2011;66:2248-2254.
by benzalkonium chloride. J Clin Microbiol. 1981;14:157-160. 31. Brachelente C, Wiener D, Malik Y, et al. A case of necrotizing fas-
19. Perez C, Fujii Y, Fauls M, et al. Fatal aortic endocarditis associated ciitis with septic shock in a cat caused by Acinetobacter baumannii. Vet
with community-acquired Serratia marcescens infection in a dog. Dermatol. 2007;18:432-438.
J Am Anim Hosp Assoc. 2011;47:133-137. 32. Weese JS, Blondeau JM, Boothe D, et al. Antimicrobial use guide-
20. Plavec T, Zdovc I, Juntes P, et al. Necrotizing fasciitis caused by lines for treatment of urinary tract disease in dogs and cats: Antimicrobial
Serratia marcescens after tooth extraction in a Doberman pinscher: Guidelines Working Group of the International Society for Companion
a case report. Veterinarni Medicina. 2008;53:629-635. Animal Infectious Diseases. Vet Med Int. 2011;2011:263768.
21. Hohenhaus AE, Drusin LM, Garvey MS. Serratia marcescens con- 33. Sugino Y, Kato M, Yagi A, et al. Pasteurella multocida pneumonia
tamination of feline whole blood in a hospital blood bank. J Am with molecular evidence of zoonotic transmission. Kansenshogaku
Vet Med Assoc. 1997;210:794-798. Zasshi. 2007;81:726-730.
22. Galarneau JR, Fortin M, Lapointe JM, et al. Citrobacter freundii
septicemia in two dogs. J Vet Diagn Invest. 2003;15:297-299.
23. Sykes JE, Kittleson MD, Pesavento PA, et al. Evaluation of the rela-
tionship between causative organisms and clinical characteristics of
infective endocarditis in dogs: 71 cases (1992-2005). J Am Vet Med
Assoc. 2006;228:1723-1734.
CHAPTER 37

Anaerobic Bacterial Infections


Jane E. Sykes

that comprise the normal flora, the anaerobic bacterial species


Overview of Anaerobic Bacterial Infections most often identified in disease are the gram-negative anaerobes
First Described: 1860s (Louis Pasteur)1 Fusobacterium spp., Bacteroides spp., Prevotella spp., and Por-
phyromonas spp., and the gram-positive anaerobes Clostridium
Causes: Bacteroides, Prevotella, Porphyromonas, Peptostrep-
spp. and Peptostreptococcus spp. (Box 37-1).2 An accurate
tococcus, Fusobacterium, Clostridium, Propionibacterium,
understanding of the epidemiology of anaerobic bacterial infec-
other species
tions in dogs and cats has been hampered by difficulties associ-
Geographic Distribution: Worldwide ated with culture of anaerobes. The use of molecular methods
Mode of Transmission: Direct contact, and opportunistic such as PCR has begun to shed more light on this topic. The
invasion of tissues by commensal bacteria reader is referred to Chapters 48 and 54 for information on
gastrointestinal Clostridium perfringens and Clostridium diffi-
Major Clinical Signs: Abscess formation in a variety of tissues,
cile infections, and disease caused by the toxins of Clostridium
pyothorax, peritonitis, pyometra, periodontal disease,
­tetani and Clostridium botulinum, respectively.
osteomyelitis, bacteremia
Differential Diagnoses: Primarily bacterial infections caused Clinical Features
by gram-positive and gram-negative aerobes, but also
neoplasia (when discrete abscessation is present). Signs and Their Pathogenesis
Human Health Significance: Anaerobes of the oral cavities of Anaerobes can cause disease in a variety of different organs.
dogs and cats are an important component of animal bite Often, their presence is associated with abscess formation and
wound infections in humans. Enteric clostridial infections necrosis at the site of inoculation, which contains a mixed popu-
of dogs and cats may also have public health significance lation of anaerobic as well as facultative bacteria. The posses-
(see Chapter 48). sion of virulence factors by anaerobes contributes to their ability
to cause disease after host barriers are disrupted. These include
capsular polysaccharides, extracellular proteases, endotoxin
(gram-negative anaerobes), and a variety of other toxins. Capsu-
Etiology and Epidemiology lar polysaccharides play an important role in the stimulation of

An anaerobe is an organism that requires an environment that is


reduced in oxygen for growth and does not grow in air. Anaer-
obic bacteria represent a significant component of the normal
BOX 37-1
flora of the upper and lower gastrointestinal tracts of dogs and Major Anaerobic Bacterial Pathogens Isolated
cats as well as their genital tracts; in addition, anaerobes play an
from Dogs and Cats
important role in the regulation of the bacterial composition of
the normal flora. Aerobes and facultative organisms that reside
alongside anaerobes use oxygen present in these sites and reduce Gram-Positive Cocci
the local oxidation-reduction potential, which permits survival Peptostreptococcus spp. (especially Peptostreptococcus
and growth of anaerobes. Sites that are protected from oxygen anaerobius)
such as the gingival crevices also favor anaerobes. The metabolic
activity of anaerobic bacteria prevents colonization of mucosal Gram-Positive Rods
surfaces by potentially more pathogenic microbes (this is known Clostridium spp.
as colonization resistance). Finally, anaerobes are important for Propionibacterium spp. (especially Propionibacterium
the maintenance of normal mucosal barrier function. acnes)
Anaerobic bacteria can cause disease when they gain access to
normally sterile sites. This occurs when mucosal barriers are dis- Gram-Negative Rods
rupted, or when anaerobes gain access to subcutaneous tissues as Bacteroides spp. (e.g., Bacteroides fragilis)
a result of a contaminated penetrating wound. Anaerobic bacte- Fusobacterium spp. (e.g., Fusobacterium necrophorum
rial infections are often mixed infections owing to the concurrent and Fusobacterium nucleatum)
presence of other anaerobes, as well as aerobes and facultative Prevotella spp. (e.g., Prevotella heparinolyticus)
bacteria such as Pasteurella spp., Escherichia coli, streptococci, Porphyromonas spp.
or staphylococci. Although there are hundreds of anaerobes

364
CHAPTER 37  Anaerobic Bacterial Infections 365

abscess formation by certain anaerobes such as Bacteroides, Por- dental plaque as a result of poor oral hygiene and other host fac-
phyromonas, Fusobacterium, and Prevotella species. Just as aer- tors. Dental plaque is a bacterial biofilm composed primarily of
obic bacteria potentiate the survival of anaerobes through local gram-positive facultative anaerobes. Subsequently there is pro-
consumption of oxygen, anaerobic bacteria produce metabolites liferation of pathogenic bacteria, which include gram-negative
such as short-chain fatty acids, which inhibit phagocytosis and rods and especially anaerobes. These secrete toxins and incite
promote survival of co-infecting facultative organisms. an inflammatory response. Anaerobes implicated in canine and
Anaerobes of the oral cavity are important contributors feline periodontal disease (such as Porphyromonas gulae) dif-
to periodontal disease, tooth root abscesses and mandibular fer from those implicated in human periodontal disease (Por-
osteomyelitis, retrobulbar abscesses, and bite wound infections. phyromonas gingivalis).3-6 Periodontal disease is manifested as
Anaerobes are frequently isolated from the lungs and pleural gingival erythema, edema, gingival bleeding, and halitosis, with
cavities of dogs or cats with aspiration pneumonia and pyotho- accumulation of calculus, gingival recession, alveolar bone loss,
rax, respectively. Intestinal anaerobes may be isolated from the and sometimes the presence of purulent exudate (Figure 37-2).
abdominal cavities of animals with septic peritonitis secondary
to intestinal perforation. The female genital tract is also heav- Retrobulbar Abscesses
ily colonized with anaerobic bacteria, and pyometra may some- Retrobulbar or orbital abscesses can result from tooth root
times be associated with mixed aerobic and anaerobic infections abscessation, penetrating foreign bodies such as grass awns, or
that include the anaerobic species Bacteroides, Prevotella, Fuso- penetrating trauma to the orbit. Clinical signs include unilateral
bacterium, and Peptostreptococcus species. exophthalmos, protrusion of the nictitans, conjunctival hyper-
Plant awn migration in dogs and cats can lead to abscess for- emia, serous to mucopurulent ocular discharge, inappetence,
mation in a variety of tissues, including retroperitoneal tissues fever, and pain on opening the mouth (Figure 37-3). A fluctuant
and the brain. These abscesses usually contain a mixed popula- swelling or draining tract posterior to the last ipsilateral molar
tion of aerobes and anaerobes, especially Fusobacterium, Acti- may be present on examination of the oral cavity. In one study,
nomyces, and Bacteroides species. Brain abscessation can follow Bacteroides spp. and Clostridium spp. were the most frequent
direct extension of anaerobic bacterial infection from otitis or anaerobes isolated from 34 dogs with retrobulbar abscesses;
tooth root infection. Alternatively, hematogenous spread may other frequently isolated bacteria were Staphylococcus spp.,
occur (Figure 37-1). Pasteurella spp., and Escherichia coli. In cats, the most frequent
anaerobe was Bacteroides vulgatus, and the most common non-
Periodontal Disease anaerobe was Pasteurella spp.7
Periodontal disease is one of the most common disorders seen
by small animal veterinarians and may result in chronic pain, Skin and Soft Tissue Infections
decreased appetite, and loss of teeth. It begins with formation of Cat bite abscesses are one of the most common reasons that cats
are brought to small animal clinics. Clinical signs include fever,
lethargy, inappetence, pain, and firm or fluctuant subcutaneous
swellings that may rupture and drain purulent material. Oral
anaerobes and Pasteurella multocida are frequently isolated
from these abscesses.8 Anaerobic species involved include Pep-
tostreptococcus anaerobius, Porphyromonas spp., Clostridium
villosum, Fusobacterium spp., Bacteroides spp. (especially Bac-
teroides tectum), and Prevotella spp.9 Despite the widespread
occurrence of cat bite abscesses, studies that use molecular
methods to identify the types and relative prevalences of anaer-
obes in cat bite abscesses are lacking.

FIGURE 37-1  Abscess in the white matter of the right frontal lobe of a 2-year-old
female spayed Chihuahua that had a 1-month history of difficulty chewing food and
lethargy and a 3-week history of progressive obtundation, neck pain, and tetrapare-
sis. Signs initially improved following treatment with prednisone, but then recurred as
the prednisone dose was reduced. The abscess in the frontal lobe communicated with
the overlying meninges, which were expanded by a chronic and fibrosing suppurative
inflammatory infiltrate. Another abscess was present in the midbrain, which suggested
hematogenous spread of infection. Although no bacteria were seen within the lesions FIGURE 37-2  Periodontitis in a dog. There is significant gingival inflammation,
with special stains, Actinomyces canis and Fusobacterium nucleatum were cultured plaque and calculus accumulation, and purulent exudate. Gingival recession and con-
from the lesions. (Courtesy of the University of California, Davis Veterinary Anatomic  tact ulceration of the buccal mucosa are also present. (Courtesy of Dr. Frank Verstraete,
Pathology Service.) University of California, Davis Dentistry and Oral Surgery Service.)
366 SECTION 2  Bacterial Diseases

Other skin and soft tissue infections associated with a variety The reader is referred to Chapter 87 for more information on
of anaerobes include foreign body penetrating wounds and dog bacterial pneumonia and pyothorax.
bite wounds (see Chapter 57).
Peritonitis
Pneumonia and Pyothorax Septic peritonitis that involves anaerobes may result from perfo-
Anaerobes such as Fusobacterium spp., Peptostreptococcus ration of the intestinal tract, especially the cecum or colon. Bac-
anaerobius, Prevotella spp., and Porphyromonas spp. are fre- teroides and Clostridium species are most often isolated from
quently isolated from dogs and cats with aspiration pneumo- these infections. Fusobacterium and Propionibacterium species
nia, foreign body pneumonia (especially that due to plant awn may also be isolated.11 Disease that results from bowel perfora-
migration), and pyothorax.10 Dogs with plant awn migration tion often contains a population of organisms that reflect the
develop multifocal necrotizing pneumonia with abscess forma- bowel flora at the site of perforation, including anaerobes, fac-
tion. In some cases, aspiration pneumonia is followed by lung ultative gram-positive anaerobes (especially Enterococcus spp.),
abscess formation (Figure 37-4). In human patients, a period Candida spp., and facultative gram-negative anaerobes such as
of 6 to 7 days is required for necrosis and abscess formation to Escherichia coli. Rupture of the female genital tract secondary
occur after aspiration, and so aspiration pneumonia with lung to pyometra may also result in anaerobic bacterial peritonitis.
abscess formation is generally a chronic disease process.
Bacteremia
Anaerobes are occasionally isolated from dogs and cats with
bloodstream infections, usually as polymicrobial infections with
aerobic bacteria. In a European study of 66 bacteremic cats,
12% of bacteria isolated from the bloodstream were obligate
anaerobes.12 In a study of 140 bacteremic dogs, anaerobes
comprised 9% of the isolates.13 Of 39 dogs and 10 cats with
critical illness and bacteremia from Colorado, anaerobes were
isolated from 31% of the dogs and 40% of the cats. The most
common anaerobe isolated from the 39 dogs was C. perfringens
(9 dogs), followed by Bacteroides species (2 dogs).14 The cats
were infected with Bacteroides or Fusobacterium species. The
possibility of mixed aerobic-anaerobic bacterial bloodstream
infection should be considered in dogs with systemic grass awn
migration. However, anaerobic bacteria appear to be a very rare
cause of endocarditis in dogs (see Chapter 86).15
A

B
FIGURE 37-3  Retrobulbar abscess and endophthalmitis secondary to a migrating
grass awn foreign body in a 10-month-old intact male Labrador retriever. A, Note the
mucopurulent ocular discharge from the right eye, as well as buphthalmos (enlarge-
ment of the eye) and exophthalmos. The nictating membrane and conjunctiva are FIGURE 37-4  Necropsy findings in an 11-year-old male neutered Labrador retriever
hyperemic, and there is diffuse corneal edema. Hypopyon and iris bombe were also with severe aspiration pneumonia secondary to laryngeal tie-back surgery for laryngeal
present. Globe retropulsion was reduced and painful on the right side. B, Multiple grass paralysis, which occurred 2 months before death. Aspiration led to bronchopneumonia
awns were present all over the dog’s haircoat, and an awn (shown) was removed from with multifocal abscessation, pleural rupture, pyothorax, and pneumothorax. There are
the gingiva posterior to the last right maxillary molar tooth. The right eye was subse- multiple to coalescing black to dark gray, soft, irregular foci in the lungs that measured up
quently enucleated. No additional grass awns were found. Histopathology of the globe to 4.5 cm in diameter. On cut surface, dark gray to black material oozed from the paren-
showed severe chronic fibrinosuppurative panophthalmitis with focal, 0.3-cm scleral chyma. Aerobic bacterial culture yielded large numbers of bacteria that included Strep-
rupture in the caudal sclera near the optic nerve, regional orbital granulation tissue, tococcus canis, nonfermenter group 3 organisms, and Pasteurella canis. Anaerobic culture
and rupture of the lens. (Courtesy of the University of California, Davis Veterinary Oph- yielded large numbers of Peptostreptococcus anaerobius. (Courtesy of the University of
thalmology Service.) California, Davis, Veterinary Anatomic Pathology Service.)
CHAPTER 37  Anaerobic Bacterial Infections 367

Diagnosis preferable to swab specimens. Specimens should immediately be


placed in an anaerobic transport medium and shipped to the
Abnormalities that suggest an anaerobic bacterial infection laboratory as soon as possible, because even brief exposure to
include a putrid odor, discolored purulent discharge, gas pro- oxygen impairs isolation. Growth is often slow when compared
duction within tissues (especially with C. perfringens infections) to other facultative organisms, and cultures should be incu-
(Figure 37-5), a bite or puncture wound, penetrating foreign bated for 7 days. Because most anaerobe infections also involve
bodies, abscessation, pyothorax, aspiration pneumonia, damage aerobes, aerobic bacterial culture should always be requested
to mucosal surfaces with which anaerobes are usually associ- concurrently.
ated, and/or necrotic or gangrenous tissue (Box 37-2). How- Extensive antimicrobial susceptibility testing is not rou-
ever, absence of these features does not rule out infection with tinely performed for anaerobes because it is laborious,
anaerobes, and some of these abnormalities occur with pure expensive, and can have a long turnaround time. Because
aerobic bacterial infections. For example, although anaerobes infections are polymicrobial (two or more anaerobe spe-
such as Clostridium spp. may be involved in emphysematous cies as well as facultative organisms present), susceptibility
cystitis in humans and dogs, the most common cause of emphy- testing for all pathogens present is often cost prohibitive to
sematous cystitis is E. coli.

Laboratory Abnormalities
BOX 37-2
Complete Blood Count, Biochemistry, and Urinalysis Findings
No specific CBC, biochemistry, or urinalysis findings suggest Disease Processes That Suggest the Possibility of Anaerobic
anaerobic infection. Infections may be associated with leukocy- Bacterial Infection
tosis due to a neutrophilia with a left shift and toxic neutrophils.
Abscess formation
Cytologic Analysis Putrid odor
Cytologic analysis of exudate in aerobic bacterial infections typ-
Necrotic tissue
ically reveals neutrophilic inflammation accompanied by bacte-
Negative cultures despite the presence of bacteria
ria of mixed morphology and gram staining characteristics. This
on cytologic examination
and/or the presence of filamentous rod-shaped bacteria within
Exposure to plant foreign bodies such as grass awns
these exudates should increase suspicion for an anaerobic infec-
Interdigital or retrobulbar soft tissue involvement
tion (Figure 37-6).
Dental infections
Microbiologic Tests Aspiration pneumonia
Pyothorax
Bacterial Isolation and Identification Gastrointestinal perforation
Optimal techniques for isolation of anaerobic bacteria are
Pyometra
described in Chapter 3. The decision to request anaerobic bac-
Bite wound infections
terial culture should be considered carefully, because anaerobes
are difficult to culture, tend to have predictable susceptibility
to antimicrobial drugs, and can contaminate specimens, which
can lead to misleading results. If anaerobic culture is performed,
the best specimens to collect are tissues or body fluids that are
normally sterile. Fluids (e.g., 1 to 2 mL) or tissue specimens are

FIGURE 37-5  Splenic abscess in an 11-year-old male neutered Labrador mix that FIGURE 37-6  Cytology of pleural fluid collected using thoracocentesis from an
also had duodenal lymphoma. Ultrasound image of a large splenic mass that contains 8-year-old female spayed domestic shorthair with pyothorax. A mixed population of
numerous hyperechoic foci and shadowing, consistent with the presence of gas. Necropsy bacteria that includes filamentous organisms suggestive of anaerobes (arrow) is present.
showed severe, focally extensive, neutrophilic, and necrotizing splenitis. The duodenum Aerobic and anaerobic bacterial culture yielded a mixed growth of Prevotella spp., Pepto-
was perforated at the site of the neoplasm. Escherichia coli, Bacteroides tectum, and a Pre- streptococcus anaerobius, Fusobacterium spp., and an unidentified anaerobic gram-positive
votella heparinolyticus–like organism were cultured from the splenic abscess. rod. Wright’s stain, 1600× magnification.
368 SECTION 2  Bacterial Diseases

TABLE 37-1
Suggested Antimicrobial Drugs for Treatment of Anaerobic Bacterial Infections in Dogs or Cats*
Drug Dose (mg/kg) Interval (hours) Route
Amoxicillin–clavulanate potassium 12.5-25 12 PO
Ampicillin-sulbactam 10-20† 8 IV, IM
Metronidazole 15 12 IV
15 12 (dogs), 24 (cats) PO
Clindamycin hydrochloride, clindamycin phosphate 11 12 (dogs), 24 (cats) PO
10 12 IV‡, IM
Chloramphenicol, chloramphenicol sodium succinate 40-50 (dogs), 12.5-20 (cats) 6-8 (dogs), 12 (cats) PO, IV, IM§
Pradofloxacin 3 (dogs), 5-10 (cats) 24 PO∥

*The concurrent presence of aerobes should also be considered when selecting antimicrobial drugs.
†Dose according to ampicillin component.
‡For IV administration, dilute 1:10 in 0.9% saline and administer over 30-60 minutes.
§See Chapter 8 for warnings about chloramphenicol use.
∥Use of pradofloxacin in dogs at high doses has been associated with myelosuppression and is extra-label in the United States. Doses shown for cats

are for the oral suspension; a dose of 3 mg/kg is recommended for the tablet formulation.

pet owners. In addition, anaerobes that infect dogs and cats a fluoroquinolone or an aminoglycoside, is a reasonable choice
generally have predictable drug susceptibilities. Neverthe- for initial treatment of serious infections while awaiting the
less, because antimicrobial resistance has increased among results of susceptibility testing for aerobic bacteria when mixed
anaerobes that infect humans, the possibility of drug resis- aerobic-anaerobic infections are suspected. For infections that
tance should be considered when infections do not respond to do not require parenteral therapy, pradofloxacin also has potent
treatment with drainage and appropriate antimicrobial drugs. activity against a variety of aerobes, anaerobes and mycoplas-
Some veterinary diagnostic laboratories perform rapid tests mas (see Chapter 8).
for β-lactamase enzyme production by anaerobes (see Case With the exception of newer generation fluoroquinolones
Example). such as pradofloxacin, anaerobes are resistant to fluoroquino-
lones. Anaerobes are also generally resistant to trimethoprim-
Treatment and Prognosis sulfamethoxazole and have unpredictable resistance to
tetracyclines. Aminoglycosides are remarkable for their com-
Treatment of anaerobic bacterial infections involves surgical plete inactivity against anaerobes.
removal of necrotic tissue, drainage of abscesses through surgi-
cal or imaging-guided approaches, restoration of blood supply, Prevention
and medical management with antimicrobial drugs (Table 37-1).
Antimicrobial drugs with activity against most anaerobes are Prevention of anaerobic bacterial infections involves manage-
β-lactam/β-lactamase inhibitor drug combinations (such as cla- ment or prevention of underlying conditions that predispose
vulanic acid–amoxicillin or ampicillin-sulbactam), clindamycin, to these infections. Vaccines for prevention of anaerobic bac-
metronidazole, chloramphenicol, and carbapenems. Although terial infections are not available. An inactivated, whole-cell
penicillin and first-generation cephalosporins may be effective, Porphyromonas denticanis, Porphyromonas gulae, and Por-
some pathogenic anaerobes that infect dogs and cats (especially phyromonas salivosa vaccine was available for several years for
Bacteroides but also a significant percentage of Prevotella, Por- prevention of periodontal disease in dogs but was withdrawn
phyromonas, and Fusobacterium isolates) may be resistant to from the market because of apparent lack of efficacy.
these drugs through the production of β-lactamase enzymes.
Approximately 20% of Bacteroides and Clostridium isolates are Public Health Aspects
resistant to clindamycin, but other anaerobes are susceptible.2
Factors such as tissue penetration should be considered when Anaerobes that reside in the oral cavity of dogs and cats are
selecting antimicrobial drugs, and the concurrent presence of important causes of bite wound infections in humans, next
aerobic bacteria must also be taken into account. Thus a combi- to Pasteurella species. The reader is referred to Chapter 57
nation of a β-lactam/β-lactamase inhibitor drug or clindamycin for more information on the public health significance of bite
and a drug with activity against gram-negative bacteria, such as wound infections.
CHAPTER 37  Anaerobic Bacterial Infections 369

CASE EXAMPLE Lymphocytes 3023 cells/µL (1000-4000 cells/µL)


Monocytes 1245 cells/µL (150-1200 cells/µL)
Platelets 392,000 platelets/µL (150,000-400,000 platelets/µL).
Signalment: “Cocoa,” a 4-month-old intact female Labrador Serum Chemistry Profile:
retriever from Gonzales in northern California Sodium 143 mmol/L (145-154 mmol/L)
History: Cocoa was brought to the University of California, Potassium 4.6 mmol/L (3.6-5.3 mmol/L)
Davis, Veterinary Medical Teaching Hospital for Chloride 102 mmol/L (108-118 mmol/L)
evaluation of seizures. Approximately 6 weeks before Bicarbonate 23 mmol/L (16-26 mmol/L)
referral, the dog was seen for left ocular exophthalmos. Phosphorus 6.9 mg/dL (3.0-6.2 mg/dL)
She was treated with amoxicillin–clavulanic acid for Calcium 11.1 mg/dL (9.7-11.5 mg/dl)
1 week, and the exophthalmos resolved, but subsequently BUN 7 mg/dL (5-21 mg/dL)
right ocular exophthalmos developed. This resolved after Creatinine 0.4 mg/dL (0.3-1.2 mg/dL)
another 1-week course of amoxicillin–clavulanic acid. Glucose 95 mg/dL (64-123 mg/dL)
One month later, Cocoa appeared painful on opening her Total protein 6.1 g/dL (5.4-7.6 g/dL)
mouth. She was treated with cephalexin and meloxicam Albumin 2.8 g/dL (3.0-4.4 g/dL)
with no apparent improvement. Two days later she became Globulin 3.3 g/dL (1.8-3.9 g/dL)
inappetent. When the owners offered her food by hand, she ALT 35 U/L (19-67 U/L)
ate, became ataxic, fell over, her thoracic limbs stiffened, AST 52 U/L (19-42 U/L)
and foamy saliva was noted around her mouth. She was ALP 103 U/L (21-170 U/L)
taken to an emergency clinic, and she vomited in the car. Creatine kinase 556 U/L (51-399 U/L)
At the emergency clinic she had three generalized seizures Gamma GT 4 U/L (0-6 U/L)
that were treated with diazepam. She was also treated with Cholesterol 262 mg/dL (135-361 mg/dL)
enrofloxacin (5 mg/kg IV, once). Total bilirubin 0.1 mg/dL (0-0.2 mg/dL).
Cocoa’s diet normally consisted of commercial dog food, and Urinalysis: SGr 1.013; pH 6.5, negative for protein, glucose,
she had been vaccinated for distemper, hepatitis, and par- ketones, bilirubin, hemoprotein, WBC, RBC, casts, and bacteria.
voviral enteritis at 8 and 12 weeks of age. There had been Imaging Findings:
no coughing, sneezing, diarrhea, exposure to ticks or toxins, Thoracic Radiographs: Cardiopulmonary structures were
trauma, or travel outside California. within normal limits.
Physical Examination: Abdominal Ultrasound: The gastric wall was mildly thickened
Body Weight: 15.3 kg with bright, echogenic speckles within the muscularis
General: Quiet but alert and responsive. Hydrated. layer. The submucosa of the stomach was echogenic. These
T = 102.8°F (39.3°C), HR = 110 beats/min, panting, mucous changes were suggestive of gastritis. The remainder of the
membranes pink, CRT = 1 second. abdominal ultrasound was unremarkable.
Eyes, Ears, Nose, and Throat: No clinically significant Spinal Radiographs: Open physes were present within the
abnormalities were noted. There was no evidence of pain vertebral column. No abnormalities were identified.
on opening the mouth provided neck movement was Brain MRI: In the precontrast brain study, a flattened, fusiform
prevented during the examination. extradural structure was identified in the dorsal part of
Integument, Musculoskeletal, Cardiovascular, Gastro­ the foramen magnum that caused moderate dorsoventral
intestinal, Genitourinary Systems, and Peripheral compression of the spinal cord (see arrow in Figure 37-7). This
Lymph Nodes: No clinically significant findings. structure was hypointense to brain but hyperintense to CSF
Neurologic Examination: Mild obtundation was noted. The on the T1-weighted images and moderately hyperintense
dog held her head down and was reluctant to move her on dual echo and fluid-attenuated inversion recovery
neck. There was moderate to marked ataxia that involved all (FLAIR) images. The postcontrast brain study showed diffuse
four limbs, with the pelvic limbs most severely affected, and meningeal enhancement that involved the spinal cord and
the right pelvic and thoracic limbs more severely affected brain, with the most prominent enhancement noted along the
than the left. Cranial nerve examination revealed only a ventral brainstem and left cerebral hemisphere. This contrast
decreased corneal reflex on the right side. Decreased placing enhancement pattern was consistent with meningitis. A
reactions were noted in all four limbs, with the right limbs discrete rim of enhancement surrounded the aforementioned
more severely affected. There was severe pain on palpation fusiform structure, which gave it a cystic appearance. The
of the cervical spine and moderate pain on palpation of the structure had partial fluid characteristics, and, in light of the
lumbosacral spine. Multifocal CNS lesions were suspected. meningeal enhancement, an abscess was suspected.
Laboratory Findings: Analysis of CSF (Lumbar CSF): CSF protein: 726 mg/dL
CBC: (reference range, <25 mg/dL)
HCT 34.4% (40%-55%) Total RBC: 5 cells/µL (reference range, 0 cells/µL)
MCV 64.9 fL (65-75 fL) Total nucleated cells: 7500 cells/µL (reference range,
MCHC 34.9 g/dL (33-36 g/dL) <2 cells/µL)
Reticulocytes 22,000 cells/µL (7000-65,000 cells/µL) Differential: 88% neutrophils, 12% large mononuclear cells
WBC 17,780 cells/µL (6000-13,000 cells/µL) Microscopic evaluation: The specimen was highly cellular
Neutrophils 12,979 cells/µL (3000-10,500 cells/µL) and contained a large number of nondegenerate neutro-
Band neutrophils 533 cells/µL phils. A few pyknotic forms were noted. Lower numbers of

Continued
370 SECTION 2  Bacterial Diseases

treatment was changed to clavulanic acid–amoxicillin (16


mg/kg PO q8h) and metronidazole (16 mg/kg PO q12h).
On day 4, Cocoa was less ataxic and had normal placing
reactions, but had a seizure while on a walk outside. The
seizure lasted 45 seconds and was manifested by jaw
chomping and defecation, but Cocoa did not fall over or lose
consciousness. Potassium bromide (40 mg/kg PO q8h for
4 days, then 40 mg/kg PO q24h) was added to the treatment
regimen, and Cocoa was discharged from the hospital.
At a recheck 3 weeks later, Cocoa’s owners reported that she had
shown steady clinical improvement and for the last week
she had been acting like a normal puppy. There had been
no seizures. She had received the final set of the puppy vac-
cine series 1 week earlier. No abnormalities were detected
on neurologic examination. A CBC showed persistent mild
nonregenerative anemia (hematocrit 34.8%), 5253 neutro-
phils/µL, and 228 band neutrophils/µL. A repeat MRI showed
a decrease in the meningeal enhancement and marked
reduction in the size of the cystic structure, although there
FIGURE 37-7  Postcontrast brain MRI, 4-month-old intact female Labrador was persistent mild spinal cord compression. Analysis of
retriever with Bacteroides fragilis meningitis and an epidural abscess located in the CSF revealed a CSF protein concentration of 37 mg/dL, <1
dorsal part of the foramen magnum. There is a flattened, fusiform structure in the RBC/µL, 24 nucleated cells/µL, with 12% neutrophils, 51%
dorsal part of the foramen magnum associated with moderate dorsoventral compres- small mononuclear cells, and 37% large mononuclear cells.
sion of the spinal cord (arrow). Diffuse meningeal enhancement and a discrete rim of Microscopic examination of the CSF revealed a heteroge-
enhancement ­surrounding the fusiform structure are present.
nous population of lymphocytes that included small mature
lymphocytes, larger reactive lymphocytes, and many plas-
­monocytoid cells, large reactive lymphocytes, and rare eo- macytoid lymphocytes along with mature plasma cells and
sinophils also were observed. Interpretation: Marked neu- moderately vacuolated macrophages. A few nondegenerate
trophilic pleocytosis (purulent inflammation). neutrophils were present. Antimicrobial drug treatment was
Cytologic Analysis of CSF (Cisternal): A direct smear continued and a recheck was recommended in 3 months,
of cisternal CSF contained disrupted cells and mildly to but the dog was subsequently lost to follow-up.
moderately degenerate neutrophils. These were admixed Comments: The history of exophthalmos increased suspicion
with low numbers of monocytoid cells and intermediate- for an anaerobic bacterial infection of the CNS in this dog
sized reactive lymphocytes. Rare neutrophils were observed and was the reason that anaerobic culture of the CSF and
with intracellular rod-shaped bacteria. Few free rod-shaped blood was performed. Given the young age of this dog, a
bacteria were noted in the background. migrating foreign body (e.g., plant awn) was suspected.
Microbiologic Testing: Aerobic urine culture: No growth The history, together with the distribution of lesions in the
was detected. CNS and the lack of lesions elsewhere, suggested direct
Aerobic and anaerobic blood cultures: No growth was detected. extension of the infection from the retrobulbar location
Aerobic and anaerobic CSF culture: Small numbers of Bacteroi- to the meninges, rather than hematogenous spread of
des fragilis, positive for β-lactamase production. infection. Because the dog was treated with antibiotics
Diagnosis: Bacteroides fragilis meningitis with extradural before culture was performed, the concurrent presence
abscess formation of other facultative organisms or other anaerobic species
Treatment and Outcome: Cocoa was treated with ticarcillin may have been obscured. The identification of β-lactamase
(50 mg/kg IV q6h), metronidazole (10 mg/kg IV q8h), and production by the B. fragilis isolate was not surprising,
crystalloid fluids (LRS with 20 mEq/L KCl at 50 mL/h, IV). because of the high (>90%) prevalence of β-lactamase
The following day her mentation improved and she became production by this species. B. fragilis is also known for its
more willing to move. On day 3 after starting antibiotics, propensity to form abscesses.

SUGGESTED READING 3. Hardham J, Reed M, Wong J, et  al. Evaluation of a monovalent


companion animal periodontal disease vaccine in an experimental
Jang SS, Breher JE, Dabaco LA, et al. Organisms isolated from dogs and mouse periodontitis model. Vaccine. 2005;23:3148-3156.
cats with anaerobic infections and susceptibility to selected antimicro- 4. Kato Y, Shirai M, Murakami M, et  al. Molecular detection of
bial agents. J Am Vet Med Assoc. 1997;210:1610-1614. human periodontal pathogens in oral swab specimens from dogs in
Japan. J Vet Dent. 2011;28:84-89.
REFERENCES 5. Perez-Salcedo L, Herrera D, Esteban-Saltiveri D, et al. Comparison
1. Sebald M, Hauser D. Pasteur, oxygen and the anaerobes revisited. of two sampling methods for microbiological evaluation of peri-
Anaerobe. 1995;1:11-16. odontal disease in cats. Vet Microbiol. 2011;149:500-503.
2. Jang SS, Breher JE, Dabaco LA, et al. Organisms isolated from dogs 6. Senhorinho GN, Nakano V, Liu C, et al. Detection of Porphyromo-
and cats with anaerobic infections and susceptibility to selected nas gulae from subgingival biofilms of dogs with and without peri-
antimicrobial agents. J Am Vet Med Assoc. 1997;210:1610-1614. odontitis. Anaerobe. 2011;17:257-258.
CHAPTER 37  Anaerobic Bacterial Infections 371

7. Wang AL, Ledbetter EC, Kern TJ. Orbital abscess bacterial isolates 12. Greiner M, Wolf G, Hartmann K. Bacteraemia in 66 cats and anti-
and in vitro antimicrobial susceptibility patterns in dogs and cats. microbial susceptibility of the isolates (1995-2004). J Feline Med
Vet Ophthalmol. 2009;12:91-96. Surg. 2007;9:404-410.
8. Love DN, Jones RF, Bailey M, et al. Isolation and characterisation 13. Greiner M, Wolf G, Hartmann K. A retrospective study of the clini-
of bacteria from abscesses in the subcutis of cats. J Med Microbiol. cal presentation of 140 dogs and 39 cats with bacteraemia. J Small
1979;12:207-212. Anim Pract. 2008;49:378-383.
9. Love DN, Malik R, Norris JM. Bacteriological warfare amongst 14. Dow SW, Curtis CR, Jones RL, et  al. Bacterial culture of blood
cats: what have we learned about cat bite infections? Vet Micro- from critically ill dogs and cats: 100 cases (1985-1987). J Am Vet
biol. 2000;74:179-193. Med Assoc. 1989;195:113-117.
10. Walker AL, Jang SS, Hirsh DC. Bacteria associated with pyotho- 15. Sykes JE, Kittleson MD, Pesavento PA, et al. Evaluation of the rela-
rax of dogs and cats: 98 cases (1989-1998). J Am Vet Med Assoc. tionship between causative organisms and clinical characteristics of
2000;216:359-363. infective endocarditis in dogs: 71 cases (1992-2005). J Am Vet Med
11. Culp WT, Zeldis TE, Reese MS, et al. Primary bacterial peritoni- Assoc. 2006;228:1723-1734.
tis in dogs and cats: 24 cases (1990-2006). J Am Vet Med Assoc.
2009;234:906-913.
CHAPTER 38

Bordetellosis
Jane E. Sykes

but B. bronchiseptica is closely related to B. pertussis and


Overview of Canine and Feline Bordetellosis B. ­parapertussis, which cause pertussis (whooping cough) in
First Described: 1911 (Bacillus bronchicanis, Ferry).1 The genus humans. In fact, there is evidence that B. pertussis evolved
Bordetella was named after Bordet, who first described from a distinct, human-associated lineage of B. bronchiseptica,4
Bordetella pertussis (Bordet and Gengou, 1906).2 and it has been recommended that B. bronchiseptica, B. per-
tussis, and B. parapertussis be reclassified as subspecies of the
Cause: Bordetella bronchiseptica, an aerobic gram-negative
“B. bronchiseptica cluster.”5 B. pertussis can colonize puppies
motile bacterium (family Alcaligenaceae)
in the absence of clinical signs of respiratory disease.6
Affected Hosts: Dogs, cats, wild carnivores, pigs, horses, rab- Worldwide, B. bronchiseptica is an important cause of respi-
bits, rodents, turkey, humans. Dogs and cats share the ratory disease in dogs and cats; it also infects and causes respi-
same strains. ratory illness in many different animal species including wild
Geographic Distribution: Worldwide carnivores, pigs, rabbits, and occasionally horses, other herbi-
vores, rodents, turkeys, and humans. There are a large number
Mode of Transmission: Aerosol, contact with contaminated
of different strains of B. bronchiseptica that vary in virulence
fomites and water sources
and host specificity. Nevertheless, the results of molecular typ-
Major Clinical Signs: Sneezing, serous to mucopurulent nasal ing efforts have shown that strains that infect dogs can be passed
discharge, harsh or honking cough (especially dogs). Dogs to cats and vice versa.7-9 The whole genome of B. bronchiseptica
and cats with bronchopneumonia can develop fever, has been sequenced.10 Its genome is larger than that of its close
lethargy, inappetence, tachypnea, productive cough, and relatives, and it possesses several plasmids, some of which medi-
mucopurulent nasal and ocular discharge. ate antimicrobial drug resistance.11
Differential Diagnoses: Upper and lower respiratory diseases Like viral respiratory infections, bordetellosis is especially
such as collapsing trachea, viral respiratory infections, prevalent in dogs and cats in shelter, pet stores, boarding
fungal pneumonia, protozoal pneumonia, respiratory facilities, and other situations where large numbers of poten-
tract neoplasia, airway foreign bodies, chronic bronchitis, tially stressed animals may have been in close contact with one
eosinophilic bronchopneumopathy or granulomatosis, another. Infections with B. bronchiseptica frequently occur in
parasitic infections (such as Filaroides, Oslerus, Capillaria, concert with respiratory viral and/or Mycoplasma spp. infec-
Paragonimus, Dirofilaria), bronchopneumonias secondary tions (see Chapters 17, 23, and 40). Unlike B. pertussis and
to conditions such as laryngeal paralysis or ciliary dyskine- B. parapertussis, B. bronchiseptica can persist in the envi-
sia, left-sided congestive heart failure ronment for at least 10 days and can grow in natural water
sources,12 but is susceptible to most disinfectants provided they
Human Health Significance: B. bronchiseptica is closely are used correctly.
related to B. pertussis and Bordetella parapertussis, which The prevalence of B. bronchiseptica infections in group-
cause whooping cough in humans. Uncommonly, it has housed animals, including shelters, varies from group to
been associated with respiratory disease in humans and group, even in the same geographic region. B. bronchiseptica
rarely systemic infections such as peritonitis, septicemia, can be isolated from apparently healthy cats and cats with
and meningitis. It primarily causes disease in the immu- respiratory disease, but is more likely to be isolated from cats
nocompromised. Contact with dogs and cats has been with respiratory disease. In one European study that included
associated with human infections, but host specificity 1748 cats from private multicat households, shelters, and
may also be strain related. breeding catteries, the more cats in the group, the greater the
chance that B. bronchiseptica would be detected. In rescue
shelters, increased seroprevalence has been associated with
poor hygiene.13 In a study of 742 cats in the United Kingdom,
Etiologic Agent and Epidemiology B. bronchiseptica was isolated from 0% of pet cats, 19.5% of
cats from rescue shelters, and 13.5% of cats from research col-
Bordetella spp. are small, pleomorphic, gram-negative cocco- onies.14 B. bronchiseptica was cultured from 3.1% of 614 cats
bacilli. They have fimbriae (pili) and can be motile by means from four shelters in Louisiana,15 5.1% of nasal swabs from
of flagella. There are at least nine different bacterial species in 59 cats with acute respiratory disease in Colorado,16 and none
the genus Bordetella (Table 38-1).3 The only species known of 22 cats with respiratory disease in a shelter in California.17
to cause disease in dogs and cats is Bordetella bronchiseptica, However, the prevalence of infection was nearly 50% in nasal

372
CHAPTER 38  Bordetellosis 373

TABLE 38-1
Species That Belong to the Genus Bordetella
Species Host(s) Disease
Bordetella Dogs, cats, pigs, Rhinitis, tracheobron-
bronchiseptica horses, rabbits, chitis, bronchopneu-
rodents, humans monia
Bordetella Humans Whooping cough
pertussis
Bordetella Humans, sheep Whooping cough
parapertussis (humans); ovine-
adapted strains
cause respiratory
disease in sheep
Bordetella Humans Upper respiratory tract FIGURE 38-1  Bordetella bronchiseptica infection. B. bronchiseptica is inhaled, adheres
holmesii disease, bacteremia to respiratory cilia by means of fimbrial adhesins, evades the immune system and secretes
Bordetella Humans Otitis media and a variety of toxins that damage the respiratory epithelium.
trematum wound infections
Bordetella avium Poultry (especially Rhinotracheitis and secretes toxins that damage the respiratory epithelium
turkey) (Figure 38-1). Adhesion of virulent strains of B. bronchiseptica
Bordetella hinzii Birds, rarely Normal flora of birds, to respiratory cilia occurs by means of fimbrial adhesins
humans bacteremia, cholan- (FIM), a filamentous hemagglutinin (FHA), pertactin, and
gitis in humans cell wall lipopolysaccharide. Bordetella colonization factor
A (BcfA) is another outer membrane protein that appears to
Bordetella petrii Environment, None, osteomyelitis be important for tracheal colonization by B. bronchiseptica.21
rarely humans B. bronchiseptica possesses genes that encode for pertussis
Bordetella Humans Bacteremia, skin infec- toxin, an important adhesin in B. pertussis. These genes are
ansorpii tion not expressed in vitro, but there is some evidence for pertussis
toxin expression in vivo.22 Evasion of host defenses is mediated
by the organism’s outer capsule (which is absent in B. pertus-
swabs from 40 cats with rhinitis from a shelter in Colorado.18 sis and B. parapertussis), adenylate cyclase toxin, and pertac-
Similarly, a high prevalence (26%) of infection was detected tin. The capsule protects it from phagocytosis and destruction
with a PCR assay in 100 cats with upper respiratory disease by complement. Adenylate cyclase toxin catalyzes the con-
from central Italy.19 version of ATP to cAMP, which inhibits the migration and
Only a few studies have examined the epidemiology of activation of phagocytes and T-lymphocytes. Pertactin allows
B. bronchiseptica infection in dogs. In a case review from the the organism to resist neutrophil-mediated clearance.23 A type
eastern United States, B. bronchiseptica was isolated from VI secretion system also appears to play a key role in per-
32 (49%) of 65 pet dogs under 1 year of age with radiographic sistence of B. bronchiseptica.24 Other virulence determinants
and microbiologic evidence of bacterial bronchopneumonia. present in the outer membrane include lipopolysaccharide
Most of these dogs had originated from breeders (40%) or pet and the Brk protein (Bordetella resistance to killing), which
stores (41%), and only 8% were from shelters. Dogs infected allow the organism to resist complement-mediated destruc-
with B. bronchiseptica were younger (7 to 35 weeks of age), had tion. At least two other important exotoxins damage respira-
been owned for a shorter period of time (median, 18 days), were tory epithelial cells and cells of the immune system, and incite
more likely to have originated from a pet store and were less cytokine production: tracheal cytotoxin (TCT) and dermone-
likely to have originated from a breeder than dogs with pneu- crotic toxin (DNT). B. bronchiseptica also possesses a type
monia caused by other organisms. B. bronchiseptica was iso- III secretion system that allows it to inject as-yet-undefined
lated from dogs with respiratory disease as well as from healthy effector proteins into cellular targets.5
dogs in a rehoming kennel in the United Kingdom.20 The inflammation and altered cell function that occur as
a result of B. bronchiseptica infection lead to increased fluid
Clinical Features and mucus secretion, and impairment in host innate immune
defenses predisposes to opportunistic viral and bacterial infec-
Signs and Their Pathogenesis tions. Clinical signs vary in severity and may reflect factors
Transmission of B. bronchiseptica occurs primarily by the such as the bacterial strain involved, host immunity, and the
airborne route, but contact with contaminated fomites and presence of co-infections. The incubation period ranges from
water sources may also be important. The organism is highly 2 to 10 days. Rhinitis and tracheobronchitis may be associated
contagious. Much of what is known about B. bronchiseptica with serous to mucopurulent nasal discharge, sneezing, stertor,
pathogenesis is based on studies of infections in host spe- and, especially in dogs, a persistent, often paroxysmal harsh
cies other than the dog or cat. B. bronchiseptica is inhaled, cough. In some dogs, development of bronchopneumonia leads
adheres to respiratory cilia, evades the immune system, to fever, productive cough, lethargy, and decreased appetite.
374 SECTION 2  Bacterial Diseases

FIGURE 38-3  Bronchoalveolar lavage cytology from a dog infected with Bordetella
bronchiseptica. Numerous coccobacilli are adhered to the cilia of columnar epithelial cells.
FIGURE 38-2  Kitten with Bordetella bronchiseptica conjunctivitis and bronchopneu- Wright stain, 1000× oil magnification. (Courtesy of Michael Scott, Michigan State Univer-
monia. The kitten was tachypneic and had mucopurulent ocular discharge. sity. In Raskin RE, Meyer D, eds. Canine and Feline Cytology: A Color Atlas and Interpretative
Guide, 2 ed, St. Louis, MO: Saunders; 2010.)

Bronchopneumonia may be due to B. bronchiseptica itself or


it may result from co-infection with other respiratory patho- Diagnostic Imaging
gens, such as canine distemper virus (CDV). In cats, infection Plain Radiography
is significantly associated with sneezing, and cough is uncom- Plain thoracic radiographs in dogs with uncomplicated borde-
mon.14 Cats with pneumonia may develop tachypnea, cyanosis, tellosis may be unremarkable or show a mild diffuse interstitial
and death. or bronchointerstitial pattern. Bordetella bronchopneumonia
Shedding of B. bronchiseptica by both dogs and cats can con- may be characterized by development of peribronchial and alve-
tinue intermittently for at least a month and sometimes several olar infiltrates and lobar consolidation.
months after infection. Evasion of the immune system through
survival inside phagocytes may explain the persistent infection Microbiologic Tests
that occurs. In contrast, B. pertussis is rapidly destroyed by The main assays used for diagnosis of bordetellosis in dogs and cats
macrophages.25 are aerobic bacterial culture and PCR assays, which can be per-
formed on nasal or oropharyngeal swabs, TTL or BAL specimens,
Physical Examination Findings or respiratory tract tissue collected at necropsy (Table 38-2). In
On physical examination, dogs with uncomplicated borde- some studies, B. bronchiseptica has been detected in nasal but not
tellosis are bright, alert, and active with a paroxysmal cough oropharyngeal swabs, but in other studies, the use of oropharyn-
that can occur throughout the examination. The cough is geal swabs was more sensitive. Collection of swab specimens from
often easily elicited on tracheal or laryngeal palpation. Con- the nasal cavity or posterior nasopharynx is preferred for diagnosis
junctivitis and serous to mucopurulent ocular and nasal dis- of pertussis in humans because these regions have ciliated epithelial
charge may be present in affected dogs and cats (Figure 38-2). cells, for which B. pertussis has affinity.27 False-negative results
Dogs and cats with complicated Bordetella bronchopneumo- occur when organism numbers in secretions are low, and depend-
nia may be pyrexic, lethargic, and tachypneic with increased ing on the sensitivity of the PCR assay used and the viability of the
respiratory effort. There may also be increased lung sounds bacteria in the specimen, culture may be positive when PCR is neg-
and/or referred upper airway noises on thoracic auscultation, ative and vice versa. Although the detection of B. bronchiseptica
and mucopurulent nasal discharge. in TTL or BAL specimens is diagnostic for bordetellosis, it may
be more difficult to interpret the significance of a positive culture
Diagnosis or PCR assay result from nasal or oropharyngeal swabs, espe-
cially among group-housed dogs and cats when the background
Laboratory Abnormalities prevalence of infection is high. Co-infections with viral respiratory
There are no specific laboratory abnormalities in uncompli- pathogens should always be considered when B. bronchiseptica is
cated bordetellosis. Usually the CBC is normal. Dogs with detected, as well as the presence of other comorbidities that sup-
bronchopneumonia can also have a normal CBC,26 or a mild press the innate or adaptive immune system that could predispose
to moderate neutrophilia, bandemia, and toxic neutrophils to bordetellosis (e.g., ciliary dyskinesia or brachycephalic obstruc-
may be present. Cytologic examination of transtracheal tive syndrome in dogs, retroviral infections in cats).
lavage (TTL) or bronchoalveolar lavage (BAL) specimens
may reveal a suppurative or mixed exudate, and some- Bacterial Culture
times intracellular and extracellular coccobacilli are visible B. bronchiseptica can usually be isolated on routine aerobic bac-
(Figure 38-3). terial culture media, such as MacConkey agar and blood agar.
CHAPTER 38  Bordetellosis 375

TABLE 38-2
Diagnostic Assays Available for Bordetellosis in Dogs and Cats
Assay Specimen Type Target Performance
Aerobic bacterial Nasal and oropharyngeal B. bronchiseptica False negatives can occur when specimens contain no
culture swabs, transtracheal and or low numbers of organisms, or after antimicrobial
bronchoalveolar lavage treatment. Allows subsequent susceptibility testing.
specimens, airway and The clinical significance of positive culture results
lung specimens collected from nasal and oropharyngeal swab specimens may be
at necropsy difficult to interpret.
PCR assays See Aerobic bacterial B. bronchiseptica Sensitivity and specificity can vary depending on assay
culture DNA design. False negatives can occur when specimens
contain no or low numbers of organisms, or after
antimicrobial treatment. Testing specimens from
multiple different anatomic sites or combining PCR
assays with other diagnostic tests can increase sensitiv-
ity. Avirulent live vaccine organisms may be detected
for several weeks after intranasal vaccination. Because
of subclinical shedding, the clinical significance of a
positive result may be difficult to interpret.
Serology Serum Antibodies against Interpretation complicated by previous vaccination
Bordetella antigens and widespread exposure.

Charcoal-based medium that is supplemented with cephalexin is to most shelters. Serology has primarily been used to study the
preferred for isolation of B. pertussis, which is more fastidious. epidemiology of infection and to examine immune responses as
For B. bronchiseptica, use of methicillin or oxacillin in selective they relate to vaccination.
media can prevent overgrowth by contaminating microflora.5
The laboratory should be informed when bordetellosis is on Pathologic Findings
the differential diagnosis list. In one study, B. bronchiseptica Gross Pathologic Findings
was isolated from cats using nasal swabs but not oropharyngeal Gross pathologic findings in dogs with uncomplicated borde-
swabs.16 The use of Dacron, rayon, or calcium alginate swabs is tellosis are usually minimal. There may be an accumulation of
preferred for culture of B. pertussis, because growth is inhibited mucopurulent exudate in the airways.
when cotton swabs are used.27 Unlike PCR assays, successful
detection of B. bronchiseptica using culture allows susceptibility Histopathologic Findings
testing, which assists in rational antimicrobial drug selection. Microscopic findings in dogs or cats with bordetellosis consist
of a suppurative inflammatory response that is usually confined
Molecular Diagnosis Using the Polymerase Chain Reaction to the ciliated portions of the respiratory tract. Cilia are mostly
Sensitive and specific real-time PCR assays that detect intact, but regional bronchial epithelial necrosis may be present.
B. bronchiseptica DNA have been described,13,28 and assays are Mild lymphoid hyperplasia may be present. Depending on the
available through several commercial veterinary diagnostic labo- stage and severity of infection, aggregates of bacteria may be
ratories. Some laboratories offer panels that also detect other observed in association with the cilia.30
respiratory pathogens, which can be useful for detection of coin-
fections. As with culture, negative results can occur when organ- Treatment and Prognosis
ism numbers are low in the specimen, or when there has been
recent antimicrobial drug use; however, PCR assays may be more Antimicrobial Treatment
likely to be positive than culture in these situations. Positive PCR B. bronchiseptica isolates from dogs and cats have the propen-
assay results can occur for at least 3 weeks after intranasal vac- sity to exhibit multidrug resistance (i.e., resistance to three or
cination.29 A conventional PCR assay that differentiates between more antimicrobial drug classes). Resistance to amoxicillin and
field strains and one vaccine strain of B. bronchiseptica has been trimethoprim-sulfamethoxazole is common. Many infections
described,29 but commercially available real-time assays that dis- are mild or self-limiting, and so antimicrobial drug treatment
tinguish between field and vaccine strains are needed. should be reserved for animals with confirmed B. bronchiseptica
infection that have persistent clinical signs (>7 to 10 days) or
Serologic Diagnosis those with more severe clinical signs and radiographic abnor-
Antibodies to B. bronchiseptica can be detected using bacte- malities that suggest bronchopneumonia. Young kittens and
rial agglutination or recombinant or whole-cell ELISA assays, puppies (<6 to 8 weeks of age) are candidates for early initiation
but serology is of limited use for diagnosis because of the high of treatment because disease in neonates may progress rapidly
prevalence of antibodies in the dog and cat population and the to pneumonia and death. Treatment should be based when-
influence of vaccination, which is routinely performed on entry ever possible on the results of culture and susceptibility testing.
376 SECTION 2  Bacterial Diseases

When susceptibility results are not available, or when they are Inadvertent parenteral administration of intranasal vaccines has
pending and bordetellosis is strongly suspected, the initial drug the potential to lead to injection site reactions, hepatic necrosis,
of choice is doxycycline. At the time of writing, doxycycline and even death in some dogs.37 Subcutaneous fluids should be
resistance is uncommon among isolates from dogs and cats,31-33 administered at the site of vaccination, and immediate treatment
and doxycycline effectively penetrates lung tissue. Unfortu- with antimicrobial drugs is indicated. It has been suggested that
nately, doxycycline is not the best choice for other secondary gentamicin sulfate solution be instilled into the affected area
bacterial pneumonias because it is bacteriostatic and parenteral (2 to 4 mg/kg gentamicin sulfate in 10 to 30 mL of saline),
doxycycline can be expensive and difficult to administer. This and dogs should then be treated with oral doxycycline for
reinforces the need for culture whenever feasible in animals 5 to 7 days.38 Special packaging has been developed by some
with evidence of pneumonia. Bacterial shedding may continue vaccine manufacturers in an attempt to prevent inadvertent par-
despite resolution of clinical signs, so affected dogs and cats enteral administration of the intranasal vaccine.
should remain in isolation from other animals whenever pos- Concerns have been raised that intranasal B. bronchisep-
sible for at least 2 months after treatment. tica vaccines may cause respiratory disease in immunosup-
Rarely, treatment with systemic antimicrobial drugs fails to pressed humans who inhale the vaccine during administration
resolve clinical signs of bordetellosis, and the organism persists or who contact vaccine organisms shed by recently immunized
despite documented susceptibility to the antimicrobial drug dogs.39,40 However, as yet, there has been no molecular proof
selected. In this situation, a short period of nebulized amino- that the B. bronchiseptica strains isolated from affected human
glycosides could be considered. Because this can cause airway patients match vaccine strains. In fact, in one report of B. bron-
irritation, its use should probably be restricted for dogs with chiseptica pneumonia in an infant that was temporally related
refractory bronchopneumonia, or severe and persistent tracheo- to intranasal vaccination of a pet dog, genetic analysis (ribotyp-
bronchitis (for example, >4 weeks in duration). ing) showed that the vaccine was not the source of the infant’s
exposure.41 The organism matched to another B. bronchiseptica
Supportive Care and Prognosis strain that had previously been isolated from a human patient.
The prognosis for dogs with uncomplicated bordetellosis is Intranasal B. bronchiseptica vaccination is followed by the
excellent. Cats and dogs with bronchopneumonia usually development of low titers of serum IgG, whereas serum IgG
require other supportive treatments, such as IV fluid therapy, responses are higher after parenteral vaccination (as might be
appropriate nutritional support, supplemental oxygen, and neb- expected). Parenteral whole-cell vaccines, and more recently
ulization (see Chapters 17 and 23). In one study, bronchopneu- acellular vaccines that include virulence factors such as pertac-
monia associated with B. bronchiseptica infection was more tin, FIM, FHA, and pertussis toxin, are widely used to prevent
severe than that associated with other bacterial agents, and pertussis in humans.26 Fewer scientific data are available on the
affected dogs also had higher PvCO2 values, were more likely to efficacy of parenteral B. bronchiseptica vaccines in dogs when
require supplemental oxygen, and had longer mean hospitaliza- compared with intranasal vaccines. One study showed that an
tion times (7.2 days compared with 4.9 days).26 intranasal vaccine was more effective than a parenteral anti-
gen extract vaccine.42 Additional challenge studies are required
Immunity and Vaccination that directly compare the efficacy of intranasal and parenteral
B. bronchiseptica vaccines for dogs.
Antibodies to outer surface molecules of B. bronchiseptica, In some countries, intranasal B. bronchiseptica vaccines are
especially IgA in mucosal secretions, are important for bacterial available for cats, which can reduce signs of disease after chal-
clearance. Several avirulent live, mucosal (intranasal or oral) lenge as long as 1 year after vaccination.43 Because disease is
B. bronchiseptica vaccines are available for dogs, some of which rarely severe in cats more than 6 weeks of age, vaccination of
also contain canine parainfluenza virus with or without canine cats has not been recommended on a routine basis,43 but could
adenovirus 2. These vaccines are designed to stimulate mucosal be considered as an adjunct to management strategies such as
immunity and provide protection within 3 days after a single reduction of overcrowding and proper disinfection if outbreaks
dose of vaccine.34 Protection with intranasal vaccines also occurs of bordetellosis are a problem in group-housed cats. Vaccina-
in the face of maternal antibody and can last at least 1 year.35,36 tion of immunocompromised cats is not recommended.44
Inactivated parenteral B. bronchiseptica vaccines are also avail-
able for dogs, but two doses administered 3 to 4 weeks apart are Prevention
required to stimulate maximal immunity, which occurs 1 week
after the second dose. The rapid onset of protection that fol- In the absence of management strategies to reduce stress, over-
lows the use of intranasal vaccines makes them the vaccine type crowding, and comorbidities, vaccination is unlikely to com-
of choice for puppies and animals introduced into heavily con- pletely prevent bordetellosis. The reader is referred to the
taminated environments, such as shelters, when immunization sections on prevention in Chapters 17 (dogs) and 23 (cats) for
in advance of entry is usually not possible. Mucosal vaccines more additional information on prevention and management of
should not be given to animals treated with antimicrobial drugs outbreaks of respiratory infections in group-housed animals.
(because they inactivate bacteria in the vaccine), and admin-
istration of intranasal vaccines to aggressive animals without Public Health Aspects
sedation may not be feasible. Transient signs of mild to moder-
ate respiratory disease occur in a small percentage of vaccinated B. bronchiseptica is an uncommon cause of respiratory disease
dogs and cats several days after mucosal vaccination. In shel- in humans, but has been isolated from humans without clini-
ters, it may not be possible to differentiate between postvaccinal cal signs and humans with mild to severe respiratory disease,
disease and infectious respiratory disease due to field pathogens, including pneumonia.45 Most, but not all, affected humans have
which complicates decisions that relate to isolation practices. underlying immunosuppressive conditions that predispose them
CHAPTER 38  Bordetellosis 377

to infection. B. bronchiseptica peritonitis has been reported in may not distinguish between B. pertussis and B. bronchiseptica.
human patients receiving continuous peritoneal dialysis.46,47 Clinically, recognition of B. bronchiseptica infections in humans
Bacteremia and meningitis have also been reported.48 Recent is important, because B. bronchiseptica is resistant to macrolides,
exposure to cats and to dogs with signs of “kennel cough” has which are first-line agents for pertussis.41 Until more is under-
been noted in most human cases,45,49 but airborne transmis- stood about the risks of canine mucosal B. bronchiseptica vac-
sion between humans can occur in hospital settings.50 At least cines for immunocompromised people, parenteral vaccination of
in mouse models, human acellular pertussis vaccines appear at-risk dogs owned by seriously immunocompromised humans
to cross-protect against B. bronchiseptica.51 Infections with is probably warranted, and the owners of these dogs should be
B. bronchiseptica may be underrecognized in people, because the instructed to avoid or minimize situations in which their dogs
direct fluorescent antibody test used for diagnosis of pertussis could come into contact with large numbers of other dogs.

CASE EXAMPLE possibly congenital. Consolidation in the area of the right


middle lung lobe was compatible with pneumonia.
Outcome: The kitten was treated with oxygen supplementation,
Signalment: 7-week-old intact female Persian kitten from terbutaline (0.01 mg/kg SC q6h), ticarcillin-clavulanate (50
California mg/kg IV q8h), LRS with 5% dextrose (1 mL/hr IV), a single
History: A research colony kitten was examined for a 3-day dose of dexamethasone (1 mg/kg IV), and a single dose of
history of respiratory signs. Three days previously, sneezing furosemide (2 mg/kg IV). Euthanasia was performed 24 hours
was observed in this kitten and one of its littermates. A later due to lack of improvement and the poor prognosis.
day later, the kitten developed lethargy, inappetence, and Necropsy Findings: An endotracheal wash was performed
respiratory distress, and the littermate was found dead. The immediately after euthanasia. This revealed marked, septic,
kitten had been treated with lactated Ringer’s solution (6 suppurative inflammation and mild to moderate epithelial
mL SC q24h) and azithromycin suspension (0.7 mg/kg PO hyperplasia. Bacteria were present in large numbers
q24h) since it became unwell. Two other kittens in the litter extracellularly, within degenerate neutrophils, and in large mats.
remained unaffected, and one had mild nasal discharge. Gross necropsy findings consisted of diffuse lung lobe collapse
All of the cats had tested negative for FeLV antigen and FIV with multiple firm dark red areas throughout the paren-
antibody using a commercially available in-house ELISA kit. chyma. On histopathology, there was severe, multi-focal
Physical Examination: to coalescing, acute necrosuppurative bronchopneumonia
Body Weight: 0.37 kg with abundant clusters of gram-negative bacteria. Exami-
General: Alert, open-mouth breathing. T = 97.5°F (36.4°C), nation of the upper respiratory tract showed mild, multifo-
HR = 220 beats/min, RR = 40 breaths/min, mucous cal neutrophilic rhinitis and mild, multifocal, lymphocytic
membranes pale pink. tracheitis. The other kitten was also necropsied, with similar
Eyes, Ears, Nose, and Throat: No abnormalities were noted. findings.
Musculoskeletal: Body condition score 4/9. Microbiologic Testing (Endotracheal Wash Specimen):
Respiratory: Markedly increased inspiratory and expiratory Culture for aerobic bacteria, Mycoplasma spp., and virus isolation
effort, tachypnea, and open-mouth breathing were noted. revealed only moderate numbers of Bordetella bronchiseptica.
Increased lung sounds were detected on auscultation. Diagnosis: Bordetella bronchiseptica bronchopneumonia.
Cardiovascular, Gastrointestinal, and Genitourinary Comments: This outbreak of severe bordetellosis in a research
Systems and Lymph Nodes: No clinically significant cat colony resulted in the death of several kittens, which
abnormalities were detected. deteriorated rapidly even in the face of supportive care.
Imaging Findings: Thoracic radiographs: There was collapse Co-infections were not detected, although PCR testing
of the left cranial lung lobe and partial volume loss in the was not performed and the negative Mycoplasma culture
left caudal lung lobe with evidence of mediastinal shift to and virus isolation results did not rule out the possibility
the left, possibly secondary to a bronchial mucus plug. A of co-infection. Although susceptibility testing was not
focal increase in opacity was noted in the area of the right performed, resistance to the antimicrobials that were used
middle lung lobe with hyperinflation of the right cranial and can occur in B. bronchiseptica. Furthermore, the dose of
caudal lung lobes. There were regions of gas trapping, and azithromycin used as reported in the medical record was
the bronchi were enlarged. Emphysema was suspected, subtherapeutic.

SUGGESTED READINGS REFERENCES


Berkelman RL. Human illness associated with use of veterinary vac- 1. Ferry NS. Further studies on the Bacillus bronchicanis, the cause of
cines. Clin Infect Dis. 2003;37:407-414. canine distemper. Am Vet Rev. 1912;16:77.
Mattoo S, Cherry JD. Molecular pathogenesis, epidemiology, and clinical 2. Bordet J, Gengou O. Le microbe de la coqueluche. Annales de
manifestations of respiratory infections due to Bordetella pertussis and L’Institut Pasteur. 1906;20:731-741.
other Bordetella subspecies. Clin Microbiol Rev. 2005;18:326-382.
378 SECTION 2  Bacterial Diseases

3. Gross R, Keidel K, Schmitt K. Resemblance and divergence: the 23. Inatsuka CS, Xu Q, Vujkovic-Cvijin I, et al. Pertactin is required
“new” members of the genus Bordetella. Med Microbiol Immunol. for Bordetella species to resist neutrophil-mediated clearance.
2010;199:155-163. Infect Immun. 2010;78:2901-2909.
4. Diavatopoulos DA, Cummings CA, Schouls LM, et  al. Bordetella 24. Weyrich LS, Rolin OY, Muse SJ, et al. A type VI secretion system
pertussis, the causative agent of whooping cough, evolved from a dis- encoding locus is required for Bordetella bronchiseptica immuno-
tinct, human-associated lineage of B. bronchiseptica. PLoS Pathog. modulation and persistence in vivo. PLoS One. 2012;7:e45892.
2005;1:e45. 25. Schneider B, Gross R, Haas A. Phagosome acidification has
5. Mattoo S, Cherry JD. Molecular pathogenesis, epidemiology, and opposite effects on intracellular survival of Bordetella pertussis
clinical manifestations of respiratory infections due to Bordetella and B. bronchiseptica. Infect Immun. 2000;68:7039-7048.
pertussis and other Bordetella subspecies. Clin Microbiol Rev. 26. Radhakrishnan A, Drobatz KJ, Culp WT, et  al. Community-
2005;18:326-382. acquired infectious pneumonia in puppies: 65 cases (1993-2002).
6. Belcher KL, Wise DJ, Veit HP. Canine colonization and transmis- J Am Vet Med Assoc. 2007;230:1493-1497.
sion of Bordetella pertussis. Ann N Y Acad Sci. 2000;916:687-690. 27. Waters V, Halperin S. Bordetella pertussis. In: Mandell GL, Ben-
7. Binns SH, Speakman AJ, Dawson S, et al. The use of pulsed-field nett JE, Dolin R, eds. Mandell, Douglas, and Bennett’s Principles
gel electrophoresis to examine the epidemiology of Bordetella and Practice of Infectious Diseases. 7th ed. Philadelphia, PA: Else-
bronchiseptica isolated from cats and other species. Epidemiol vier; 2010:2955-2964.
Infect. 1998;120:201-208. 28. Koidl C, Bozic M, Burmeister A, et  al. Detection and differen-
8. Dawson S, Jones D, McCracken CM, et al. Bordetella bronchisep- tiation of Bordetella spp. by real-time PCR. J Clin Microbiol.
tica infection in cats following contact with infected dogs. Vet Rec. 2007;45:347-350.
2000;146:46-48. 29. Iemura R, Tsukatani R, Micallef MJ, et al. Simultaneous analysis of
9. Foley JE, Rand C, Bannasch MJ, et al. Molecular epidemiology of the nasal shedding kinetics of field and vaccine strains of Bordetella
feline bordetellosis in two animal shelters in California. USA. Prev bronchiseptica. Vet Rec. 2009;165:747-751.
Vet Med. 2002;54:141-156. 30. Bemis DA, Greisen HA, Appel MJ. Pathogenesis of canine borde-
10. Parkhill J, Sebaihia M, Preston A, et al. Comparative analysis of the tellosis. J Infect Dis. 1977;135:753-762.
genome sequences of Bordetella pertussis, Bordetella parapertussis 31. Speakman AJ, Binns SH, Dawson S, et al. Antimicrobial susceptibility
and Bordetella bronchiseptica. Nat Genet. 2003;35:32-40. of Bordetella bronchiseptica isolates from cats and a comparison of
11. Speakman AJ, Binns SH, Osborn AM, et  al. Characterization of the agar dilution and E-test methods. Vet Microbiol. 1997;54:63-72.
antibiotic resistance plasmids from Bordetella bronchiseptica. 32. Speakman AJ, Dawson S, Corkill JE, et al. Antibiotic susceptibil-
J Antimicrob Chemother. 1997;40:811-816. ity of canine Bordetella bronchiseptica isolates. Vet Microbiol.
12. Kirilenko NI. Survival of Bordetella pertussis in the air and on some 2000;71:193-200.
objects. Zh Mikrobiol Epidemiol Immunobiol. 1965;42:39-42. 33. Johnson LR, Queen EV, Vernau W, et al. Microbiologic and cyto-
13. Helps CR, Lait P, Damhuis A, et  al. Factors associated with logic assessment of bronchoalveolar lavage fluid from dogs with
upper respiratory tract disease caused by feline herpesvirus, lower respiratory infection: 105 cases (2001-2011), 2012 (Submit-
feline calicivirus, Chlamydophila felis and Bordetella bronchi- ted for publication).
septica in cats: experience from 218 European catteries. Vet Rec. 34. Gore T, Headley M, Laris R, et al. Intranasal kennel cough vaccine
2005;156:669-673. protecting dogs from experimental Bordetella bronchiseptica chal-
14. Binns SH, Dawson S, Speakman AJ, et  al. Prevalence and risk lenge within 72 hours. Vet Rec. 2005;156:482-483.
factors for feline Bordetella bronchiseptica infection. Vet Rec. 35. Lehar C, Jayappa H, Erskine J, et al. Demonstration of 1-year dura-
1999;144:575-580. tion of immunity for attenuated Bordetella bronchiseptica vaccines
15. Hoskins JD, Williams J, Roy AF, et al. Isolation and characteriza- in dogs. Vet Ther. 2008;9:257-262.
tion of Bordetella bronchiseptica from cats in southern Louisiana. 36. Jacobs AA, Theelen RP, Jaspers R, et  al. Protection of dogs for
Vet Immunol Immunopathol. 1998;65:173-176. 13 months against Bordetella bronchiseptica and canine parainflu-
16. Veir JK, Ruch-Gallie R, Spindel ME, et al. Prevalence of selected enza virus with a modified live vaccine. Vet Rec. 2005;157:19-23.
infectious organisms and comparison of two anatomic sampling 37. Toshach K, Jackson MW, Dubielzig RR. Hepatocellular necrosis
sites in shelter cats with upper respiratory tract disease. J Feline associated with the subcutaneous injection of an intranasal Borde-
Med Surg. 2008;10:551-557. tella bronchiseptica-canine parainfluenza vaccine. J Am Anim Hosp
17. Burns RE, Wagner DC, Leutenegger CM, et  al. Histologic and Assoc. 1997;33:126-128.
molecular correlation in shelter cats with acute upper respiratory 38. Larson LJ, Newbury S, Schultz R. Canine and feline vaccinations
infection. J Clin Microbiol. 2011;49:2454-2460. and immunology. In: Miller L, Hurley K, eds. Infectious Disease
18. Spindel ME, Veir JK, Radecki SV, et  al. Evaluation of prado- Management in Animal Shelters. Ames, IA: Wiley-Blackwell;
floxacin for the treatment of feline rhinitis. J Feline Med Surg. 2009:61-82.
2008;10:472-479. 39. Gisel JJ, Brumble LM, Johnson MM. Bordetella bronchiseptica
19. Di Martino B, Di Francesco CE, Meridiani I, et al. Etiological inves- pneumonia in a kidney-pancreas transplant patient after exposure
tigation of multiple respiratory infections in cats. New Microbiol. to recently vaccinated dogs. Transpl Infect Dis. 2010;12:73-76.
2007;30:455-461. 40. Berkelman RL. Human illness associated with use of veterinary
20. Chalker VJ, Toomey C, Opperman S, et  al. Respiratory disease vaccines. Clin Infect Dis. 2003;37:407-414.
in kennelled dogs: serological responses to Bordetella bronchi- 41. Rath BA, Register KB, Wall J, et  al. Persistent Bordetella bron-
septica lipopolysaccharide do not correlate with bacterial isola- chiseptica pneumonia in an immunocompetent infant and genetic
tion or clinical respiratory symptoms. Clin Diagn Lab Immunol. comparison of clinical isolates with kennel cough vaccine strains.
2003;10:352-356. Clin Infect Dis. 2008;46:905-908.
21. Sukumar N, Mishra M, Sloan GP, et  al. Differential Bvg phase- 42. Davis R, Jayappa H, Abdelmagid OY, et  al. Comparison of the
dependent regulation and combinatorial role in pathogen- mucosal immune response in dogs vaccinated with either an intrana-
esis of two Bordetella paralogs, BipA and BcfA. J Bacteriol. sal avirulent live culture or a subcutaneous antigen extract vaccine of
2007;189:3695-3704. Bordetella bronchiseptica. Vet Ther. 2007;8:32-40.
22. Stefanelli P, Mastrantonio P, Hausman SZ, et al. Molecular char- 43. Williams J, Laris R, Gray AW, et  al. Studies of the efficacy of a
acterization of two Bordetella bronchiseptica strains isolated from novel intranasal vaccine against feline bordetellosis. Vet Rec.
children with coughs. J Clin Microbiol. 1997;35:1550-1555. 2002;150:439-442.
CHAPTER 38  Bordetellosis 379

44. Egberink H, Addie D, Belak S, et  al. Bordetella bronchiseptica 49. Goldberg JD, Kamboj M, Ford R, et al. “Kennel cough” in a patient
infection in cats. ABCD guidelines on prevention and management. following allogeneic hematopoietic stem cell transplant. Bone Mar-
J Feline Med Surg. 2009;11:610-614. row Transplant. 2009;44:381-382.
45. Wernli D, Emonet S, Schrenzel J, et  al. Evaluation of eight cases 50. Huebner ES, Christman B, Dummer S, et  al. Hospital-acquired
of confirmed Bordetella bronchiseptica infection and colonization Bordetella bronchiseptica infection following hematopoietic stem cell
over a 15-year period. Clin Microbiol Infect. 2011;17:201-203. transplantation. J Clin Microbiol. 2006;44:2581-2583.
46. Hadley K, Torres AM, Moran J, et  al. Bordetella bronchiseptica 51. Goebel EM, Zhang X, Harvill ET. Bordetella pertussis infection or
peritonitis—beware of the dog! Perit Dial Int. 2009;29:670-671. vaccination substantially protects mice against B. bronchiseptica
47. Byrd LH, Anama L, Gutkin M, et  al. Bordetella bronchiseptica infection. PLoS One. 2009;4:e6778.
peritonitis associated with continuous ambulatory peritoneal dialy-
sis. J Clin Microbiol. 1981;14:232-233.
48. Woolfrey BF, Moody JA. Human infections associated with Borde-
tella bronchiseptica. Clin Microbiol Rev. 1991;4:243-255.
CHAPTER 39

Cell Wall–Deficient Bacterial Infections


Jane E. Sykes

Clinical Features
Overview of Cell Wall–Deficient Bacterial
Infections In human patients, CWDB have been implicated in culture-­
First Described: 1935, London (Kleineberger)1 negative endocarditis, bacteremia, uveitis, pneumonia, osteomy-
elitis and arthritis, urinary tract infections, soft tissue infections,
Causes: Cell wall–deficient strains of a variety of gram-positive
and meningitis.3 In cats, CWDB have been implicated as a cause
and gram-negative bacteria
of subcutaneous abscesses and arthritis.4,5 A cell wall–deficient
Geographic Distribution: Worldwide Nocardia sp. was isolated from a dog with polyarthritis that
Major Clinical Signs: Chronic draining or ulcerated soft tissue was refractory to antibiotics,6 and a Pseudomonas aeruginosa
and skin lesions, polyarthritis, possibly other chronic pyo- L-form was isolated from the blood of a dog after antibiotic
genic and pyogranulomatous lesions treatment for P. aeruginosa endocarditis.7
Differential Diagnoses: Infections due to other fastidious
bacteria such as Mycoplasma, Nocardia, Mycobacterium,
Diagnosis
Bartonella, Brucella, nutritionally variant streptococci such Physical examination and laboratory findings in cats and dogs
as Granulicatella, or, for animals with polyarthritis, Bor- infected with CWDB may be similar to those for other chronic,
relia burgdorferi; fungal infections (e.g., due to Sporothrix persistent bacterial infections. Infection with CWDB should be
schenckii or Coccidioides immitis); persistence of a foreign considered in cats and dogs with culture-negative pyogranulo-
body; neoplasia; congenital immunodeficiency; primary matous or suppurative inflammatory lesions, or when a bacte-
immune-mediated disease rial etiology is suspected on the basis of clinical signs but routine
Human Health Significance: Likely similar to that for the cor-
responding walled (or “normal”) variant of the bacterial
species involved
BOX 39-1
Examples of Medically Important Bacteria
That May Become Cell Wall–Deficient When Subjected
to Appropriate Stressors
Etiologic Agent and Epidemiology
Staphylococcus spp.
Cell wall–deficient bacteria (CWDB), also known as L-phase Streptococcus spp.
or L-form bacteria, are bacterial variants that lack a cell wall, Enterococcus spp.
although they may in fact possess small amounts of peptido- Escherichia coli
glycan.2 The name L-form was given to these bacteria because Serratia marcescens
they were discovered at the Lister Institute in London. L-form Pasteurella multocida
bacteria are distinct from mycoplasmas, because Mycoplasma Pseudomonas aeruginosa
spp. do not originate from bacteria that normally possess a Proteus mirabilis
cell wall. A huge variety of gram-­positive and gram-negative Salmonella typhimurium
bacterial species may become CWDB when exposed to cer- Lactobacillus spp.
tain stressors in the laboratory (such as antimicrobial drugs) Nocardia spp.
(Box 39-1). Some of these bacteria remain as CWDB (stable Actinomyces spp.
L-forms), whereas others revert back to possession of a cell Bacillus spp.
wall (unstable L-forms). CWDB assume a spherical or pleo- Brucella spp.
morphic shape, and are susceptible to osmotic lysis. How- Corynebacterium spp.
ever, they resist β-lactam drugs such as penicillin, and may be Borrelia burgdorferi
able to evade the innate immune response. Thus, it has been Leptospira interrogans
proposed that reversion to cell wall–deficient forms may be a
mechanism of bacterial persistence. Although the formation Adapted from Onwuamaegbu ME, Belcher RA, Soare C. Cell wall-
of CWDB has been well documented in the laboratory, the deficient bacteria as a cause of infections: a review of the clinical
role that CWDB play in disease remains controversial.3 significance. J Int Med Res 2005;33:1-20.

380
CHAPTER 39  Cell Wall–Deficient Bacterial Infections 381

TABLE 39-1
Assays Available for Diagnosis of Cell Wall–Deficient Bacterial Infections in Dogs and Cats
Assay Specimen Type Target Performance
Culture Biopsies, body fluids, Cell wall–deficient Requires special L-form bacterial (hyperosmolar)
wash specimens bacteria (CWDB) media
Cytology or Biopsies, smears of CWDB Organisms usually not visible owing to the lack of a
­histopathology affected tissues cell wall, but may be gram-variable. Low sensitiv-
ity and cannot be used to identify absence of a
cell wall (although the presence of organisms that
stain strongly with Gram stain indicates the pres-
ence of walled bacteria).
Electron microscopy Biopsies CWDB Confirms the presence of CWDB in lesions. May not
distinguish CWDB from Mycoplasma spp. Often
expensive and not readily available for routine
diagnostic purposes.
Broad-range bacte- Biopsies, body fluids, 16S ribosomal subunit Confirms presence of bacteria in lesions when
rial PCR assay wash specimens DNA of CWDB culture is negative. False positives may occur if
and sequencing contaminating bacteria are present. Does not
distinguish between CWDB and their walled
counterparts. Sensitivity and specificity may vary
between assays.

cultures are negative. Definitive diagnosis is difficult because the SUGGESTED READINGS
organisms are not always readily visualized using light micros- Carro T, Pedersen NC, Beaman BL, et al. Subcutaneous abscesses and
copy, and passage through special hypertonic laboratory media arthritis caused by a probable bacterial L-form in cats. J Am Vet Med
is required for growth of CWDB in culture (Table 39-1). Elec- Assoc. 1989;194:1583-1588.
tron microscopy has been used to identify the organisms in Chmel H. Graft infection and bacteremia with a tolerant L-form of
lesions. The use of broad-range bacterial PCR assays that detect Streptococcus sanguis in a patient receiving hemodialysis. J Clin
bacterial 16S ribosomal subunit DNA in tissues may be use- Microbiol. 1986;24:294-295.
ful to detect L-form bacteria in tissues in the future when rou-
tine culture methods are negative, although this method does REFERENCES
not distinguish between CWDB and their walled counterparts. 1. Kleineberger E. The natural occurrence of pleuropneumonia-like
Infection with CWDB could also be suspected when lesions organisms in apparent symbiosis with Streptobacillus moniliformis
resolve after treatment with tetracyclines. and other bacteria. J Pathol Bacteriol. 1935;40:93-105.
2. Joseleau-Petit D, Liebart JC, Ayala JA, et  al. Unstable Escherichia
coli L forms revisited: growth requires peptidoglycan synthesis.
Treatment and Prognosis J Bacteriol. 2007;189:6512-6520.
3. Onwuamaegbu ME, Belcher RA, Soare C. Cell wall–deficient bacte-
CWDB are generally susceptible to tetracyclines such as ria as a cause of infections: a review of the clinical significance. J Int
doxy­cycline. Other antimicrobial drugs that act through cell Med Res. 2005;33:1-20.
wall–­independent mechanisms, such as fluoroquinolones, chlor- 4. Carro T, Pedersen NC, Beaman BL, et  al. Subcutaneous abscesses
amphenicol, and macrolides, may also be active against some and arthritis caused by a probable bacterial L-form in cats. J Am Vet
L-form bacteria. Med Assoc. 1989;194:1583-1588.
5. Keane DP. Chronic abscesses in cats associated with an organism
Public Health Aspects resembling Mycoplasma. Can Vet J. 1983;24:289-291.
6. Buchanan AM, Beaman BL, Pedersen NC, et al. Nocardia asteroides
recovery from a dog with steroid- and antibiotic-unresponsive idio-
The role that CWDB play in zoonotic disease is unclear, but it
pathic polyarthritis. J Clin Microbiol. 1983;18:702-708.
is reasonable to believe it is similar to that of the corresponding 7. Bone WJ. L-form of Pseudomonas aeruginosa the etiologic agent
walled (or normal) variant of the bacterial species implicated. A of bacterial endocarditis in a dog. Vet Med Small Anim Clin.
single report described isolation of an L-form of Streptococcus 1970;65:224-227.
sanguinis from a vascular graft of a hemodialysis patient.8 The 8. Chmel H. Graft infection and bacteremia with a tolerant L-form
affected person’s pet dog was reported to often lick the graft site, of Streptococcus sanguis in a patient receiving hemodialysis. J Clin
and S. sanguinis was also isolated from the dog’s oral cavity. Microbiol. 1986;24:294-295.
Although this was suggestive of zoonotic transmission, the iden-
tity of the isolates was not confirmed with molecular methods.
CHAPTER 40

Mycoplasma Infections
Jane E. Sykes

Overview of Mycoplasma and Ureaplasma BOX 40-1


Infections Nonhemotropic Mycoplasma and Ureaplasma Species Found
First Described: Dogs, Japan 1934 (Shoetensack).1 Reports of
in Dogs and Cats3-7
human Mycoplasma infections also occurred around this
time.2
Dogs Cats
Causes: Mycoplasma cynos and Mycoplasma felis have been M. cynos M. felis
most commonly associated with disease, but many other M. canis M. gateae
Mycoplasma species infect dogs and cats M. molare M. arginini
Geographic Distribution: Worldwide M. arginini M. feliminutum
M. bovigenitalium M. canadense*
Mode of Transmission: Direct contact, fomite transmission
M. edwardii M. cynos*
in overcrowded environments. Opportunistic invasion of
M. feliminutum M. lipophilum*
commensal mycoplasmas also occurs.
M. felis M. hyopharyngis*
Major Clinical Signs: Conjunctivitis, sneezing and nasal dis- M. gateae U. felinum
charge (cats); cough, tachypnea, fever, weight loss (lower M. maculosum U. cati
respiratory disease); swollen, painful joints (polyarthritis); M. opalescens Acholeplasma laidlawii
subcutaneous abscesses; neurologic signs (meningoen- M. spumans
cephalitis); prostatitis, epididymitis-orchitis, or lower uri- M. sp. HRC689
nary tract signs (dogs) M. sp. VJC358
Differential Diagnoses: Primarily gram-positive and gram- U. canigenitalium
negative bacterial infections. Differential diagnoses for Acholeplasma laidlawii
suspected mycoplasma respiratory disease include viral
infections, fungal and parasitic causes of pneumonia, aspi- *Identified on the basis of DNA sequencing of small segments of the
ration pneumonia, and neoplasia. 16S rRNA gene; sequence analysis of longer fragments may reveal
Human Health Significance: Canine and feline mycoplasma these are in fact other mycoplasma species.
species have rarely been detected in immunocompro-
mised humans with mycoplasmosis who have had con-
successfully cultured in the laboratory (see Chapter 41). The
tact with pet cats and dogs.
remainder of this chapter refers only to nonhemotropic myco-
plasma species.
Mycoplasmas are commensal bacteria found widely in asso-
ciation with mucous membranes of all mammalian species.
Etiology and Epidemiology Simultaneous colonization with more than one Mycoplasma
species is common. Mycoplasmas have also been associated
Mycoplasmas are fastidious bacteria that lack a cell wall. They with keratoconjunctivitis and/or upper respiratory tract disease
belong to the class Mollicutes (which translates to “soft skin”), (URTD) in cats; reproductive disease and urinary tract infec-
and are the smallest known free-living organisms. Many require tions (UTIs) in dogs; and lower respiratory tract disease, skin
sterols for growth, and Ureaplasma species require urea for fer- and soft tissue infections, meningoencephalitis, and arthritis in
mentation. Mycoplasmas measure only 0.3 to 0.8 µm in size. both cats and dogs (Box 40-2).8-18 Because mycoplasmas are
More than 120 different species of Mycoplasma and 7 species of commonly isolated from the upper respiratory and genital tracts
Ureaplasma have been identified. The spectrum of mycoplasma of healthy dogs and cats, their role in ocular, upper respiratory
species that infect or colonize dogs and cats is incompletely tract, and urogenital tract disease has been difficult to deter-
understood because precise species identification has been dif- mine. Several studies have now identified an increased preva-
ficult (Box 40-1).3-7 Additional mycoplasma species are likely lence of mycoplasmas in cats with conjunctivitis or URTD when
to be discovered with the application of molecular methods in compared with healthy cats.13,19,20 Mycoplasmas can act as
the future. Some Mycoplasma spp. infect multiple host species. secondary invaders when other pathogenic viruses and bacteria
Mycoplasmas are often difficult to grow on cell-free media, and are present, or when normal host defenses are impaired by fac-
some, such as the hemotropic mycoplasmas, have never been tors such as underlying neoplasia or immunosuppressive drug

382
CHAPTER 40  Mycoplasma Infections 383

BOX 40-2
Diseases That May Be Associated with Mycoplasma
Infections in Dogs and Cats

Cats
Conjunctivitis
Upper respiratory tract disease
Pyothorax

Dogs
Urinary tract infections
Prostatitis
Epididymitis and orchitis
FIGURE 40-1  Ulcerative keratitis in a 4-year-old female spayed Persian. Involvement
Both Cats and Dogs of feline herpesvirus-1 was suspected, and Mycoplasma spp. was isolated from a corneal
Bronchopneumonia swab. The significance of the Mycoplasma infection was unclear, but the ulcer healed after
Skin and soft tissue infections (abscesses) topical treatment with topical ofloxacin and azithromycin drops. (Courtesy of the Univer-
Meningoencephalitis sity of California, Davis, Veterinary Ophthalmology Service.)
Polyarthritis

Conjunctivitis and Upper Respiratory Tract Disease


Mycoplasma spp. are commonly detected in cats in associa-
therapy. When mycoplasmas are associated with systemic dis- tion with conjunctivitis and URTD. Concurrent infections with
ease or pneumonia, young animals (less than 1 year of age) are other upper respiratory pathogens (such as Bordetella bron-
often involved. Some mycoplasma species appear to be more chiseptica, Chlamydia felis, and feline respiratory viruses) may
pathogenic than others. For example, evidence has accumulated contribute to clinical signs. Stressors such as overcrowding and
that Mycoplasma cynos is associated with lower respiratory dis- unhygienic conditions may promote proliferation of mycoplas-
ease in dogs.21,22 In one study, M. cynos infection was associated mas and their transmission between cats. Ulcerative keratitis has
with increased severity of canine infectious respiratory disease, been reported in association with Mycoplasma infection, but the
younger age, and longer time spent in a kennel.22 Experimen- presence of corneal involvement may reflect the concurrent pres-
tal infection with M. cynos leads to respiratory disease in dogs, ence of feline herpesvirus 1 (FHV-1) or other factors that predis-
but infection with Mycoplasma gateae, Mycoplasma canis, and pose to secondary invasion by Mycoplasma spp. (Figure 40-1).
Mycoplasma spumans does not.23 The role of mycoplasmas in
feline lower urinary tract disease and lymphoplasmacytic podo- Pneumonia and Pyothorax
dermatitis has been investigated using molecular techniques, but Virtually all dogs and cats harbor mycoplasmas in their upper
no association was found.24,25 respiratory tracts. Fewer than 25% of healthy dogs harbor
mycoplasmas in their trachea or lungs. In one study myco-
Clinical Features plasmas were detected in tracheobronchial lavage specimens
from 21% of 28 cats with pulmonary disease, but not from
Signs and Their Pathogenesis 18 healthy cats.27,28 Mycoplasmas can be isolated from the
In order to colonize mammalian hosts, mycoplasmas must lower airways and lungs of dogs and cats with pneumonia,
adhere to host cells. Mycoplasma species that infect humans are sometimes in pure culture.29 Factors that predispose to the
known to possess specific adhesins, but these have not been well development of mycoplasma pneumonia are not always appar-
characterized in mycoplasmas from dogs and cats. Invasion of ent.30 Clinical signs include variable fever, cough, tachypnea,
deeper tissues and disease occurs as a result of immunosuppres- lethargy, and decreased appetite. Concurrent signs of URTD
sion or disruption of normal host barriers. Some mycoplasmas, have been reported in some affected cats.30 Rarely, mycoplas-
such as the human pathogen Mycoplasma pneumoniae, pro- mas can be cultured from the pleural fluid of cats and dogs
duce toxins, but whether similar virulence factors are present in with pyothorax, in pure culture or in mixed infections with
mycoplasmas isolated from dogs and cats is unknown. A num- other organisms.31-33
ber of pathogenic mycoplasma species of humans can invade
cells, which may contribute to organism persistence and dif- Urogenital Tract
ficulties associated with isolation in the laboratory. Transmis- Mycoplasma and Ureaplasma spp. have been found in the semen
sion of pathogenic mycoplasmas such as M. cynos may occur in and urine of dogs with infertility and purulent epididymitis.34
overcrowded environments as a result of close contact or fomite The role of mycoplasmas in reproductive tract disease in dogs
spread. The finding of identical strains in multiple dogs from a and cats is unclear because they are commonly isolated from
kennel environment (using molecular typing methods) supports the urogenital tract of healthy dogs and cats. Large numbers
the concept of transmission between dogs in this environment.26 of mycoplasmas (>105 CFU/mL) can occasionally be isolated
The extent to which mycoplasmas survive in the environment in pure culture from urine collected by cystocentesis from dogs
varies among mycoplasma species and has not been determined with prostatitis and lower urinary tract signs.14,34 Dogs with
for mycoplasmas that infect dogs and cats. mycoplasma UTIs often have other comorbidities that impair
384 SECTION 2  Bacterial Diseases

normal urinary defenses, such as urinary tract neoplasia, calculi,


or neurologic disease.14

Arthritis and Bacteremia


Isolated case reports exist of polyarthritis due to infection with
M. spumans or Mycoplasma edwardii (dogs) and M. gateae or
M. felis (cats). Synovial fluid analysis usually reveals the pres-
ence of large numbers of neutrophils. This may lead to diag-
nostic confusion with primary immune-mediated polyarthritis.
Systemic signs such as lethargy, fever, and inappetence usually
accompany lameness. Evidence of immune compromise, such
as underlying neoplasia or surgery, young age, or a history of
glucocorticoid treatment, has generally been present in dogs and
cats with mycoplasma polyarthritis. M. felis has also been iso-
lated from cats with monoarthritis, possibly secondary to bite
wound infections.17

Meningoencephalitis
Mycoplasmas may gain access to the central nervous system
after penetrating wounds, ascending infection from the middle
ear, or hematogenous spread. Meningoencephalitis associated
with M. felis infection was described in a 10-month-old cat that
showed signs of lethargy, fever, a head tilt, nystagmus, and tet-
raparesis.12 M. felis was isolated in pure culture from the CSF.
The cat also had evidence of otitis media-interna at necropsy,
but the role this played in development of the mycoplasma
infection was unclear. M. edwardii was isolated from the brain
FIGURE 40-2  Ventrodorsal thoracic radiograph from a 3-month-old intact female
of a 6-week-old dog with a sudden onset of seizures.8 Suppu- pug with Mycoplasma cynos pneumonia. There is consolidation of the lung lobes associ-
rative and histiocytic meningitis was detected at necropsy, in ated with the left hemithorax and the right middle lung lobe. A diffuse interstitial pattern
conjunction with evidence of a possible penetrating wound to is present in the remainder of the right lung fields.
the skull. Recently, infection with Mycoplasma canis has been
associated with some cases of granulomatous meningoencepha-
lomyelitis and necrotizing meningoencephalitis in dogs,35 which Diagnostic Imaging
requires further study. Plain Radiography
Mycoplasma pneumonia may be characterized by interstitial
Physical Examination Findings to alveolar patterns or lung lobe consolidation (Figure 40-2);
Physical examination findings in dogs and cats with mycoplasma sometimes, mild pleural effusion is present. Radiographs of
infections depend on the organ affected, as well as underlying the joints of dogs and cats with mycoplasma polyarthritis may
disease processes that facilitate opportunistic invasion by myco- reveal periarticular soft tissue swelling. Erosive changes to the
plasmas. Ocular infections in cats are characterized by serous subchondral bone have also been described.9
to mucopurulent ocular discharge, conjunctival hyperemia, and
possibly keratitis. Upper respiratory tract infections may result Microbiologic Tests
in nasal discharge and sneezing, with or without conjunctivi- Assays available for diagnosis of mycoplasma infections in dogs
tis. Fever, tachypnea, and increased lung sounds may be present and cats are shown in Table 40-1.
with pneumonia. Urinary tract infections may lead to clinical
signs of lower urinary tract disease, such as hematuria or stran- Cytologic Examination
guria. Mycoplasma arthritis may be manifested by fever, joint Mycoplasma spp. are often not visible by light microscopy
pain and swelling, and sometimes local lymphadenopathy.9-11 because of their small size. They do not stain with Gram stain
because they lack a cell wall. Cytologic examination of affected
Diagnosis body fluids (such as bronchoalveolar lavage fluid, synovial fluid,
and CSF) usually reveals a predominance of neutrophils with
Laboratory Abnormalities fewer histiocytes and lymphocytes.9,10,12,29,30
Complete Blood Count, Serum Biochemical Tests, and Urinalysis
Findings on the CBC, serum biochemistry panel, and urinaly- Isolation and Identification
sis in dogs or cats with Mycoplasma infections are nonspecific Specimens suitable for mycoplasma culture include synovial
and influenced by underlying immunocompromising disease fluid, blood, CSF, urine collected by cystocentesis, pleural fluid,
processes. Dogs and cats with pneumonia, mycoplasma bac- semen, and tracheobronchial or bronchoalveolar lavage speci-
teremia, and polyarthritis may have a neutrophilia with a left mens. Swabs of the nasal cavity, conjunctiva, cornea, urethra,
shift and neutrophil toxicity, a degenerative left shift, mild vagina, or cervix can also be submitted, but interpretation of
nonregenerative anemia, hypoalbuminemia, and evidence of results from these sites is difficult. If swabs are used, the swab
organ dysfunction such as increased liver enzyme activities and should be rubbed vigorously on the mucosa, because mycoplas-
azotemia.9,10,15 mas are cell associated. Rapid transport to the laboratory is
CHAPTER 40  Mycoplasma Infections 385

TABLE 40-1
Assays Available for Diagnosis of Mycoplasma Infections in Dogs and Cats
Assay Specimen Type Target Performance
Culture Swabs, body fluids, lavage Cultivable mycoplasma Some organisms grow on blood agar, but others require
specimens, tissue obtained species special mycoplasma media. The presence of unculti-
at necropsy vable mycoplasmas may lead to false-negative results.
Turnaround time may be slow (several days to weeks);
species differentiation and antimicrobial drug suscep-
tibility testing is often not performed. The significance
of positive results from the upper respiratory and
genital tract may be unclear.
PCR assays Swabs, body fluids, lavage Mycoplasma DNA Allows rapid detection of mycoplasmas. More sensitive
specimens, tissue obtained than culture for detection of fastidious or uncultivable
at necropsy mycoplasmas, although sensitivity and specificity may
vary between assays. Some assays are designed to only
detect certain pathogenic Mycoplasma species, such as
Mycoplasma cynos. Sequencing of the PCR product
may permit species identification. The clinical signifi-
cance of positive results may be unclear.

important, because mycoplasmas are susceptible to deteriora-


tion in the environment. The use of special transport media (e.g.,
Stuart’s medium or Amies medium without charcoal) for swab
or tissue specimens may increase yield of fastidious mycoplasma
species if a delay of several hours is likely between collection of
specimens and transport to the laboratory. Specimens should be
refrigerated if delayed transport is unavoidable. If the delay is
likely to be longer than 24 hours, specimens in transport media
should ideally be frozen at −80°C. Some mycoplasma species
will still grow in the laboratory if specimens are kept refriger-
ated for 2 to 3 days.
Some mycoplasmas grow on blood agar under routine aero-
bic or anaerobic conditions. Others require special mycoplasma
media, and some fail to grow under any laboratory conditions. FIGURE 40-3  Colonies of Mycoplasma bovirhinis showing the “fried egg” appear-
The rate of growth in culture varies from one Mycoplasma spe- ance that is typical of the growth of some Mycoplasma spp. in culture. (From The genera
cies to another; for some mycoplasmas, colonies are visible in Mycoplasma and Ureaplasma. In Songer JG Post KW, eds. Veterinary Microbiology Bacterial
2 or 3 days, whereas others require several weeks of incuba- and Fungal Agents of Animal Disease, 2 ed, St. Louis, MO: Saunders; 2005.)
tion before colonies appear. Examination of plates under the
light microscope may be required for detection of Mycoplasma
colonies, because the colonies range in size from 15 to 300 µm Antimicrobial susceptibility testing for mycoplasmas is labor
in diameter. The colonies of many mycoplasma species have a intensive and not routinely performed, and established break-
characteristic “fried egg” appearance (Figure 40-3). Biochemi- points are not available. When performed, the most widely used
cal properties can then be used to subgroup mycoplasmas, but method is microbroth dilution (see Chapter 3).36
species identification has traditionally required the use of spe-
cies-specific antisera. Because cross-reactions can occur between Molecular Diagnosis Using the Polymerase Chain Reaction
species even when antisera are used, molecular typing methods Some veterinary diagnostic laboratories offer PCR assays that
(such as 16S rRNA gene or 16S/23S rRNA intergenic spacer directly detect mycoplasma DNA in clinical specimens, without
region PCR assays and sequence analysis of the respective PCR the need for culture. Several conventional and real-time PCR
products) are now the method of choice for species identifica- assays have been used to detect mycoplasmas of dogs and
tion.3 Most veterinary diagnostic laboratories currently report cats.37-40 These assays may be specific for certain mycoplasma
only growth of a Mycoplasma species and do not identify species (such as M. cynos or M. felis) or they may be genus-
organisms to the species level unless specifically requested. It is specific assays that detect a variety of mycoplasma species.
difficult to interpret the significance of a positive mycoplasma PCR assays may be more sensitive than culture for detection of
culture from mucosal sites that can normally be colonized by fastidious mycoplasma species. Sequencing of the PCR prod-
mycoplasmas, including the lower respiratory tracts of dogs, so uct generated from genus-specific PCR assays may be used to
results must be interpreted in light of the clinical findings and identify the infecting species present, provided the PCR ampli-
the results of other diagnostic tests. con is sufficiently long. As with culture, the significance of a
386 SECTION 2  Bacterial Diseases

TABLE 40-2
Suggested Antimicrobial Drug Treatment for Mycoplasma
Infections in Dogs and Cats
Drug Dose (mg/kg) Route Interval (hours)
Doxycycline 5 to 10 PO or IV 12 to 24
Enrofloxacin* 5 PO or IV 24
Marbofloxacin 2.75 to 5.5 PO 24
Pradofloxacin† 5 to 10 (cats) PO 24
3 mg/kg (dogs)

*For cats, use enrofloxacin only when other alternatives are not possible
because of risk for irreversible blindness.
†Use of pradofloxacin is extra-label in dogs in the United States (see

Chapter 8). The dose for cats is for the oral suspension. The dose for the
tablet formulation is 3 mg/kg.
A

Treatment and Prognosis


Treatment of mycoplasma infections involves specific antimi-
crobial therapy (Table 40-2) as well as management of other
underlying disorders that predisposed to invasion by mycoplas-
mas. Tetracyclines or fluoroquinolones are active against most
mycoplasma isolates. The optimal duration is unknown, but
treatment for at least 2 weeks is reasonable. In a prospective
study of shelter cats with URTD and M. felis infection, a 14-day
course of doxycycline was more effective in causing reduction
in M. felis load as determined with real-time PCR than a 7-day
course, but there was no overall significant difference in the
reduction in clinical signs between the two groups.41 Neverthe-
less, 25% of the cats treated for 14 days were still infected after
that time. In another study of cats with conjunctivitis, three
B weeks of treatment with pradofloxacin or doxycycline was
required to eliminate Mycoplasma DNA.40 β-Lactam drugs are
FIGURE 40-4  Histopathology of the inguinal subcutaneous tissues (A) and not active against mycoplasmas, because they lack any pepti-
sublumbar lymph node (B) from a 6-month-old, retrovirus–negative, male neu- doglycan. Mycoplasmas are also intrinsically resistant to trime-
tered domestic shorthair with severe suppurative cellulitis, steatitis, dermatitis, and
thoprim-sulfonamide combinations and rifampin. Some species
myositis of the scrotal skin and subcutaneous tissue. A pure culture of a Mycoplasma
species was obtained from the lesion. The cat had been neutered 13 days previously are resistant to macrolides.
and was evaluated for a 2-day history of lethargy and reluctance to move. Physical
examination revealed fever (104.6°F [40.3°C]), conjunctivitis, generalized peripheral Immunity and Vaccination
lymphadenomegaly, and firm mass lesions in the inguinal area. In addition to suppu-
rative cellulitis and myositis, marked systemic lymph-node hyperplasia was present. Mycoplasma spp. persist in tissues in the face of the immune
Hyperplasia was most profound in the sublumbar lymph nodes, which were 4 cm in response. Mechanisms to evade the host immune response such
diameter. as antigenic variation of outer surface proteins have been identi-
fied in several mycoplasma species. Innate, humoral (especially
IgA), and cell-mediated immune responses may be required to
positive PCR test result for Mycoplasma spp. may be unclear clear mycoplasma infections, with humoral mechanisms of criti-
given the role the frequent colonization of healthy animals cal importance. Vaccines to limit disease caused by pathogenic
with mycoplasmas, so results must be interpreted with the mycoplasma species such as M. cynos are currently not avail-
underlying disease process and the results of other diagnostic able, although mycoplasma vaccines have been used with success
tests. in food production animal species.

Pathologic Findings Prevention


Gross pathology in dogs and cats with mycoplasmal diseases
usually reveals purulent material in affected tissues. Histo- Prevention of mycoplasma infections involves management or
pathology shows predominantly neutrophilic inflammation, reversal of disorders that predispose to opportunistic invasion
although histiocytes may also be present. Reactive lymphade- by mycoplasmas. Reduction of overcrowding and concurrent
nopathy may also be evident as a result of immune stimulation infections in shelter environments may also reduce the inci-
(Figure 40-4). dence and severity of mycoplasma infections (see Chapter 11).
CHAPTER 40  Mycoplasma Infections 387

Routine disinfectants are sufficient to inactivate mycoplasmas woman had worked in an animal shelter, lived with a cat, and
that persist in the environment. had been bitten on the hand by a healthy cat 6 months before
she was evaluated. A mycoplasma was also isolated from a
Public Health Aspects cat scratch wound on the hand of a veterinarian.44 Although
Staphylococcus aureus was initially isolated from the wound,
The vast majority of mycoplasma species associated with dis- it failed to resolve completely after treatment with erythro-
ease in humans are different from those found in dogs and mycin. Subsequently, tenosynovitis developed, and a myco-
cats. However, pharyngeal colonization with M. canis was plasma was isolated. The identity of the mycoplasma could
reported in a family and their pet dog.42 Mycoplasma felis not be determined using biochemical and serologic methods,
was isolated from the joints of a woman with common vari- and whether a cat was truly the source of this infection was
able immunodeficiency who developed septic arthritis.43 The not proven.

CASE EXAMPLE CBC:


HCT 35.4% (40%-55%)
MCV 68.6 fL (65-75 fL)
Signalment: “Delilah”, a 3-month old intact female pug dog MCHC 31.1 g/dL (33-36 g/dL)
from Reno, NV WBC 31,580 cells/µL (6000-13,000 cells/µL)
History: Delilah was brought to the University of California, Neutrophils 6948 cells/µL (3000-10,500 cells/µL)
Davis, Veterinary Medical Teaching Hospital for evaluation Band neutrophils 9790 cells/µL
of pneumonia. She had been purchased from a pet store 3 Lymphocytes 9158 cells/µL (1000-4000 cells/µL)
weeks before the date of evaluation. The owner noticed nasal Monocytes 4737 cells/µL (150-1200 cells/µL)
and ocular discharge when the dog was brought home from Eosinophils 947 cells/µL (150-1100 cells/µL)
the store, as well as a dry, nonproductive cough. Delilah was Platelets 307,000 platelets/µL (150,000-400,000 platelets/µL).
taken to a local veterinary clinic where a diagnosis of “kennel Band neutrophils and mature neutrophils had evidence of
cough” was made. She was then treated with clavulanic acid– toxic changes.
amoxicillin (15 mg/kg PO q12h) for 2 weeks. However, the Serum Chemistry Profile:
cough persisted, and Delilah’s respiratory effort increased. She Sodium 147 mmol/L (145-154 mmol/L)
was hospitalized at the local veterinary clinic and treated with Potassium 4.9 mmol/L (3.6-5.3 mmol/L)
ticarcillin-clavulanate (50 mg/kg IV q8h), supplemental oxygen, Chloride 104 mmol/L (108-118 mmol/L)
nebulization, and coupage. Radiographs showed infiltrates in Bicarbonate 24 mmol/L (16-26 mmol/L)
the left lung lobes and right middle lung lobe that worsened Phosphorus 8.0 mg/dL (3.0-6.2 mg/dL)
over the course of hospitalization, so she was referred for Calcium 10.5 mg/dL (9.7-11.5 mg/dl)
further evaluation and treatment. There had been no decrease BUN 10 mg/dL (5-21 mg/dL)
in appetite, vomiting, or diarrhea. She was fed a commercial Creatinine 0.5 mg/dL (0.3-1.2 mg/dL)
puppy food and had no history of ticks, travel, trauma, or toxin Glucose 110 mg/dL (64-123 mg/dL)
exposure. The owner was not aware of previous vaccinations. Total protein 5.9 g/dL (5.4-7.6 g/dL)
Physical Examination: Albumin 2.5 g/dL (3.0-4.4 g/dL)
Body Weight: 2.2 kg Globulin 3.4 g/dL (1.8-3.9 g/dL)
General: Quiet, alert, hydrated. T = 101.0°F (38.3°C), HR = 220 ALT 12 U/L (19-67 U/L)
beats/min, RR = 60 breaths/min, slightly tacky mucous AST 38 U/L (19-42 U/L)
membranes with a normal capillary refill time. ALP 144 U/L (21-170 U/L)
Eyes, Ears, Nose, and Throat: Mild serous oculonasal Gamma GT 0 U/L (0-6 U/L)
discharge was present bilaterally. Conjunctival hyperemia Cholesterol 188 mg/dL (135-361 mg/dL)
and chemosis were present, as well as scleral injection. Total bilirubin 0.0 mg/dL (0-0.2 mg/dL).
Musculoskeletal: BCS 4/9. Fully ambulatory with no Urinalysis: SGr 1.045; pH 7.0, no protein, glucose, ketones,
evidence of muscle atrophy. Poor body conformation was bilirubin, WBC, or RBC were present. Rare triple phosphate
present, characterized by medial deviation of the tarsi and and a few sodium urate crystals were noted.
hyperextension of the carpi. Imaging Findings:
Respiratory: Tachypnea was present with marked abdominal Thoracic Radiographs: There was complete consolidation
effort. Lung sounds were markedly increased, especially of the lung lobes associated with the left hemithorax (see
on the left side. There were no crackles or wheezes. Figure 40-2). Prominent air bronchogram formation was
A nonproductive cough occurred several times during the noted in these lobes and in the right middle lung lobe, which
examination. also appeared consolidated. Diffuse interstitial markings
All Other Systems: No clinically significant abnormalities were were present in the remainder of the right lung fields.
detected. Thoracic Ultrasound: Complete consolidation of the left lung
Laboratory Findings: was the only abnormality detected.
Venous Blood Gas: pH = 7.348, pCO2 44 mm Hg, pO2 44.7 mm Cytologic Findings: Transtracheal lavage: One direct smear
Hg, HCO3 23.2 mmol/L, base excess −1.4 mmol/L, ionized was evaluated that was highly cellular and contained a minimal
calcium 1.43 mmol/L, lactate 3.3 mmol/L. amount of mucus. Cells were essentially all neutrophils,

Continued
388 SECTION 2  Bacterial Diseases

which were primarily nondegenerate but occasionally mildly enrofloxacin, amoxicillin-clavulanate, coupage, and exercise
to moderately degenerate. A single extracellular rod and restriction was continued at home. At a recheck 3 weeks later,
two neutrophils with intracellular structures that resembled Delilah had gained weight (3.5 kg). An intermittent, moist,
bacteria were found. Interpretation: Marked suppurative but nonproductive cough was present only in the morning.
inflammation. Her appetite was normal, and there was no evidence of
Microbiologic Testing: Gram-stained direct smear of exercise intolerance or oculonasal discharge. A physical
transtracheal lavage fluid: Large numbers of neutrophils examination was unremarkable. A CBC showed persistent
were seen. mild regenerative anemia, with 7260 neutrophils/µL and 330
Aerobic and anaerobic bacterial culture and Mycoplasma cul- band neutrophils/µL. Thoracic radiographs showed improved
ture (transtracheal wash fluid): Large numbers of Mycoplas- aeration of the left lung lobes, and near complete resolution
ma spp.; DNA sequence analysis revealed 100% identity to of the right-sided infiltrates. The amoxicillin-clavulanate
Mycoplasma cynos. No other aerobic or anaerobic organisms was discontinued. Further improvement in the cough and
were isolated. pulmonary infiltrates was observed at a recheck 3 weeks
Direct fluorescent antibody for distemper virus antigen (con- later. After an additional 7 weeks of treatment, the cough had
junctival smear): Negative resolved, but partial atelectasis of the cranial portion of the
Centrifugal zinc sulfate fecal flotation: Negative left lung lobe persisted.
Diagnosis: Mycoplasma cynos bronchopneumonia Comments: The isolation of a pure culture of Mycoplasma
Treatment: Before the culture results were available, Delilah cynos from this dog’s airways, the lack of response to
was treated with supplemental oxygen, nebulization, β-lactam antibiotics, and a prompt response to enrofloxacin
coupage, and IV fluids. Treatment with ticarcillin-clavulanate treatment supported a diagnosis of mycoplasma pneumonia
was continued for 3 days, then changed to enrofloxacin (and possibly also URTD) in this dog. The concurrent
(5 mg/kg PO q24h) and amoxicillin-clavulanate (19 mg/kg PO presence of respiratory viruses remains possible because
q12h). Daily nebulization with gentamicin was also initiated, other than direct fluorescent antibody testing for canine
because there had been no improvement and persistent distemper virus, diagnostic tests for respiratory viruses were
bordetellosis was considered possible. The following morning, not performed, and co-infection with multiple respiratory
lung sounds were improved and Delilah was playful. Her nasal pathogens in dogs from pet stores and shelter environments
discharge resolved and the frequency of cough reduced. The is common. Resolution of the cough and radiographic
CBC showed a regenerative anemia (hematocrit 33.2%, with changes occurred over several months. Concern existed
95,300 reticulocytes); 14,112 slightly toxic neutrophils/µL; in regard to cartilage toxicity in such a young dog due to
1008 band neutrophils/µL; 5796 lymphocytes/µL; and prolonged treatment with enrofloxacin, but no adverse
3780 monocytes/µL. Growth of Mycoplasma spp. was effects occurred. The regenerative anemia was interesting
reported 5 days after specimens were submitted for culture. and may have reflected pulmonary hemorrhage or immune-
Oxygen supplementation was withdrawn, and Delilah was mediated hemolysis. Reticulocytosis has been reported in
discharged on day 8 of hospitalization, although significant human patients with community-acquired pneumonia due
radiographic improvement was not yet evident and an to Mycoplasma pneumoniae.
occasional productive cough was still present. Treatment with

SUGGESTED READINGS 5. Heyward JT, Sabry MZ, Dowdle WR. Characterization of myco-
plasma species of feline origin. Am J Vet Res. 1969;30:615-622.
Chalker VJ, Owen WM, Paterson C, et  al. Mycoplasmas associated 6. Tan RJ. Susceptibility of kittens to Mycoplasma felis infection.
with canine infectious respiratory disease. Microbiology. 2004;150: Jpn J Exp Med. 1974;44:235-240.
3491-3497. 7. Chalker VJ. Canine mycoplasmas. Res Vet Sci. 2005;79:1-8.
Hartmann AD, Hawley J, Werckenthin C, et  al. Detection of bacterial 8. Ilha MR, Rajeev S, Watson C, et al. Meningoencephalitis caused by
and viral organisms from the conjunctiva of cats with conjunctivitis and Mycoplasma edwardii in a dog. J Vet Diagn Invest. 2010;22:805-808.
upper respiratory tract disease. J Feline Med Surg. 2010;12:775-782. 9. Zeugswetter F, Hittmair KM, de Arespacochaga AG, et al. Erosive
Stenske KA, Bemis DA, Hill K, et al. Acute polyarthritis and septicemia polyarthritis associated with Mycoplasma gateae in a cat. J Feline
from Mycoplasma edwardii after surgical removal of bilateral adrenal Med Surg. 2007;9:226-231.
tumors in a dog. J Vet Intern Med. 2005;19:768-771. 10. Stenske KA, Bemis DA, Hill K, et al. Acute polyarthritis and septi-
cemia from Mycoplasma edwardii after surgical removal of bilat-
REFERENCES eral adrenal tumors in a dog. J Vet Intern Med. 2005;19:768-771.
1. Shoetensack HM. Pure cultivation of filterable virus isolated from 11. Barton MD, Ireland L, Kirschner JL, et  al. Isolation of Myco-
canine distemper. Kitasako Archives Exp Med. 1934;11:227-290. plasma spumans from polyarthritis in a greyhound. Aust Vet J.
2. Baseman JB, Tully JG. Mycoplasmas: sophisticated, reemerg- 1985;62:206-207.
ing, and burdened by their notoriety. Emerg Infect Dis. 12. Beauchamp DJ, da Costa RC, Premanandan C, et al. Mycoplasma
1997;3(1):21-32. felis–associated meningoencephalomyelitis in a cat. J Feline Med
3. Spergser J, Rosengarten R. Identification and differentiation of Surg. 2011;13:139-143.
canine Mycoplasma isolates by 16S-23S rDNA PCR-RFLP. Vet 13. Haesebrouck F, Devriese LA, van Rijssen B, et al. Incidence and sig-
Microbiol. 2007;125:170-174. nificance of isolation of Mycoplasma felis from conjunctival swabs
4. Hartmann AD, Hawley J, Werckenthin C, et al. Detection of bac- of cats. Vet Microbiol. 1991;26:95-101.
terial and viral organisms from the conjunctiva of cats with con- 14. Jang SS, Ling GV, Yamamoto R, et  al. Mycoplasma as a
junctivitis and upper respiratory tract disease. J Feline Med Surg. cause of canine urinary tract infection. J Am Vet Med Assoc.
2010;12:775-782. 1984;185:45-47.
CHAPTER 40  Mycoplasma Infections 389

15. Moise NS, Crissman JW, Fairbrother JF, et al. Mycoplasma gateae 30. Foster SF, Barrs VR, Martin P, et  al. Pneumonia associated with
arthritis and tenosynovitis in cats: case report and experimental Mycoplasma spp. in three cats. Aust Vet J. 1998;76:460-464.
reproduction of the disease. Am J Vet Res. 1983;44:16-21. 31. Malik R, Love DN, Hunt GB, et  al. Pyothorax associated with
16. Hooper PT, Ireland LA, Carter A. Mycoplasma polyarthritis in a cat Mycoplasma species in a kitten. J Small Anim Pract. 1991;32:
with probable severe immune deficiency. Aust Vet J. 1985;62:352. 31-34.
17. Liehmann L, Degasperi B, Spergser J, et al. Mycoplasma felis arthri- 32. Gulbahar MY, Gurturk K. Pyothorax associated with a Myco-
tis in two cats. J Small Anim Pract. 2006;47:476-479. plasma sp. and Arcanobacterium pyogenes in a kitten. Aust Vet J.
18. Gray LD, Ketring KL, Tang YW. Clinical use of 16S rRNA 2002;80:344-345.
gene sequencing to identify Mycoplasma felis and M. gateae 33. Walker AL, Jang SS, Hirsh DC. Bacteria associated with pyotho-
associated with feline ulcerative keratitis. J Clin Microbiol. rax of dogs and cats: 98 cases (1989-1998). J Am Vet Med Assoc.
2005;43:3431-3434. 2000;216:359-363.
19. Low HC, Powell CC, Veir JK, et al. Prevalence of feline herpesvirus 34. L’Abee-Lund TM, Heiene R, Friis NF, et  al. Mycoplasma canis
1, Chlamydophila felis, and Mycoplasma spp DNA in conjunctival and urogenital disease in dogs in Norway. Vet Rec. 2003;153:
cells collected from cats with and without conjunctivitis. Am J Vet 231-235.
Res. 2007;68:643-648. 35. Barber RM, Porter BF, Li Q, et  al. Broadly reactive polymerase
20. Holst BS, Hanas S, Berndtsson LT, et al. Infectious causes for feline chain reaction for pathogen detection in canine granulomatous
upper respiratory tract disease—a case-control study. J Feline Med meningoencephalomyelitis and necrotizing meningoencephalitis.
Surg. 2010;12:783-789. J Vet Intern Med. 2012;26:962-968.
21. Rycroft AN, Tsounakou E, Chalker V. Serological evidence of 36. Waites KB, Taylor-Robinson D. Mycoplasma and Ureaplasma.
Mycoplasma cynos infection in canine infectious respiratory dis- In: Versalovic J, Carroll KC, Funke G, Jorgensen JH, Landry ML,
ease. Vet Microbiol. 2007;120:358-362. Warnock DW, eds. Manual of Clinical Microbiology. Washington,
22. Chalker VJ, Owen WM, Paterson C, et  al. Mycoplasmas associ- DC: ASM Press; 2011:970-985.
ated with canine infectious respiratory disease. Microbiology. 37. Rebelo AR, Parker L, Cai HY. Use of high-resolution melting
2004;150:3491-3497. curve analysis to identify Mycoplasma species commonly isolated
23. Rosendal S. Canine mycoplasmas: pathogenicity of myco- from ruminant, avian, and canine samples. J Vet Diagn Invest.
plasmas associated with distemper pneumonia. J Infect Dis. 2011;23:932-936.
1978;138:203-210. 38. Soderlund R, Bolske G, Holst BS, et al. Development and evalu-
24. Abou N, Houwers DJ, van Dongen AM. PCR-based detection ation of a real-time polymerase chain reaction method for the
reveals no causative role for Mycoplasma and Ureaplasma in feline detection of Mycoplasma felis. J Vet Diagn Invest. 2011;23:
lower urinary tract disease. Vet Microbiol. 2006;116:246-247. 890-893.
25. Bettenay SV, Lappin MR, Mueller RS. An immunohistochemical 39. Chalker VJ, Owen WM, Paterson CJ, et  al. Development of a
and polymerase chain reaction evaluation of feline plasmacytic polymerase chain reaction for the detection of Mycoplasma felis in
pododermatitis. Vet Pathol. 2007;44:80-83. domestic cats. Vet Microbiol. 2004;100:77-82.
26. Mannering SA, McAuliffe L, Lawes JR, et  al. Strain typing of 40. Hartmann AD, Helps CR, Lappin MR, et  al. Efficacy of prado-
Mycoplasma cynos isolates from dogs with respiratory disease. Vet floxacin in cats with feline upper respiratory tract disease due to
Microbiol. 2009;135:292-296. Chlamydophila felis or Mycoplasma infections. J Vet Intern Med.
27. Randolph JF, Moise NS, Scarlett JM, et  al. Prevalence of myco- 2008;22:44-52.
plasmal and ureaplasmal recovery from tracheobronchial lavages 41. Kompare B, Litster AL, Leutenegger CM, et  al. Randomized
and of mycoplasmal recovery from pharyngeal swab speci- masked clinical trial to compare 7-day and 14-day course length of
mens in cats with or without pulmonary disease. Am J Vet Res. doxycycline in the treatment of Mycoplasma felis infection in shel-
1993;54:897-900. ter cats. Comp Immunol Microbiol Infect Dis. 2012:Dec 13 [Epub].
28. Randolph JF, Moise NS, Scarlett JM, et al. Prevalence of mycoplas- 42. Armstrong D, Yu BH, Yagoda A, et al. Colonization of humans by
mal and ureaplasmal recovery from tracheobronchial lavages and Mycoplasma canis. J Infect Dis. 1971;124:607-609.
prevalence of mycoplasmal recovery from pharyngeal swab speci- 43. Bonilla HF, Chenoweth CE, Tully JG, et al. Mycoplasma felis sep-
mens in dogs with or without pulmonary disease. Am J Vet Res. tic arthritis in a patient with hypogammaglobulinemia. Clin Infect Dis.
1993;54:387-391. 1997;24:222-225.
29. Chandler JC, Lappin MR. Mycoplasmal respiratory infections in 44. McCabe SJ, Murray JF, Ruhnke HL, et al. Mycoplasma infection of
small animals: 17 cases (1988-1999). J Am Anim Hosp Assoc. the hand acquired from a cat. J Hand Surg Am. 1987;12:1085-1088.
2002;38:111-119.
CHAPTER 41

Hemoplasma Infections
Jane E. Sykes and Séverine Tasker

mycoplasmal pathogens Mycoplasma pneumoniae and Myco-


Overview of Hemoplasma Infections plasma genitalium.
First Described: Feline hemoplasmas, South Africa, 1942 At least three hemoplasma species infect domestic as well as
(Clark)1; canine hemoplasmas, Germany, 1928 (Kikuth)2 wild cats, Mycoplasma haemofelis, ‘Candidatus Mycoplasma
haemominutum,’ and ‘Candidatus Mycoplasma turicensis.’ The
Cause: Mycoplasma haemofelis, ‘Candidatus Mycoplasma
‘Candidatus’ prefix is applied to newly discovered hemoplasmas
haemominutum,’ ‘Candidatus Mycoplasma turicensis’
until more information is available to support their classifica-
(cats). The most prevalent hemoplasmas in dogs are
tion. This is because hemoplasmas cannot be cultured in the
Mycoplasma haemocanis and ‘Candidatus Mycoplasma
laboratory, which limits full characterization of these organ-
haematoparvum.’
isms. M. haemofelis (previously the Ohio strain, or large form
Affected Hosts: Cats and wild felids, dogs and wild canids; of Haemobartonella felis) is the most pathogenic organism and
other hemoplasma species cause disease in a variety of can cause moderate to severe hemolytic anemia in immunocom-
other animal species. petent cats. The resulting disease has been referred to as feline
Geographic Distribution: Worldwide infectious anemia. Using cytologic evaluation of blood smears,
M. haemofelis organisms are cocci that sometimes form short
Mode of Transmission: Experimentally, transmission can
chains of three to six organisms (Figure 41-1). M. haemofelis is
occur through ingestion or injection of infected blood;
the least prevalent of the three feline hemoplasmas. It has been
the mode of transmission in the field uncertain. Biting or
found using PCR in 0.5% to 5% of sick cats at veterinary hos-
aggressive interactions are a suspected mode, and pos-
pitals. The whole genome sequences of M. haemofelis and M.
sibly vertical transmission. Ticks are suspected to transmit
haemominutum have been determined.3-5
Mycoplasma haemocanis. Other unidentified arthropod
‘Ca. M. haemominutum’ (previously the California strain or
vectors may also be involved.
small form of H. felis) is generally smaller than M. haemofelis
Major Clinical Signs: Fever, lethargy, inappetence, weakness, and has not clearly been associated with disease in immunocom-
pallor. petent cats. Using cytologic evaluation of blood smears, ‘Ca.
Differential Diagnoses: Cytauxzoonosis, FIV infection, FeLV M. haemominutum’ are small cocci, 0.3 to 0.6 µm in diameter,
infection, feline infectious peritonitis, primary immune- although M. haemofelis and ‘Ca. M. haemominutum’ cannot
mediated hemolytic anemia, Heinz body hemolytic ane-
mia, inherited erythrocyte disorders such as pyruvate
kinase deficiency and the red cell fragility disorder of
Abyssinian and Somali cats
Human Health Significance: DNA sequences that match
those found in Mycoplasma haemofelis were detected in a
human co-infected with HIV and Bartonella henselae. DNA
sequences of other animal hemoplasmas have also been
detected in humans.

Etiology and Epidemiology


Hemotropic mycoplasmas (hemoplasmas) are small (0.3-0.8
µm), unculturable mycoplasmas that reside on the surface of
erythrocytes and can cause variable degrees of hemolytic anemia
in infected hosts. Hemoplasmas infect a wide variety of mam-
malian species, including humans, and have a worldwide distri-
bution. Although previously classified as Haemobartonella and
Eperythrozoon spp., sequence analysis of the 16S rRNA genes FIGURE 41-1  Romanowsky-stained blood smear from an 8-year-old male neutered
of these organisms has shown that they are closely related to the domestic shorthair cat showing epierythrocytic bacteria typical of Mycoplasma haemofelis
pneumoniae group of mycoplasmas, which includes the human (arrows).

390
CHAPTER 41  Hemoplasma Infections 391

always reliably be distinguished by this method. ‘Ca. M. haemo- of acquiring persistent subclinical infection increases over time.
minutum’ is common in the cat population worldwide, infect- In contrast, young cats may be more likely to develop disease
ing as many as 20% of cats that visit veterinary hospitals.6-8 after infection by M. haemofelis. Some studies, but not others,
Infection of cats with ‘Ca. M. haemominutum’ results in a mild have shown an association between retrovirus and hemoplasma
decrease in hematocrit. There is some evidence that ‘Ca. M. hae- infections. Cats infected with M. haemofelis in the United States
mominutum’ may play a role in disease. For example, cats that were 6 times more likely to be FIV infected than cats negative
are co-infected with FeLV and ‘Ca. M. haemominutum’ develop for hemoplasmas.15
more significant anemia than cats infected with ‘Ca. M. hae- Several hemoplasma species also infect dogs. Infection with
mominutum’ alone, and progression to FeLV-induced myelo- Mycoplasma haemocanis (previously Haemobartonella canis) has
proliferative disease occurred more rapidly.9 There are also been associated with hemolytic anemia in splenectomized dogs,
descriptions of cats with hemolytic anemia in which the only and rarely in dogs with other immunosuppressive disease or con-
recognized cause of anemia was ‘Ca. M. haemominutum.’10 ‘Ca. current infections. The 16S rRNA gene of M. haemocanis has the
M. haemominutum’ is commonly found in co-infections with same sequence as M. haemofelis, but the whole genome sequence
‘Ca. M. turicensis’ or M. haemofelis. Mixed infections with all of M. haemocanis distinguishes it as a different species.17 M. hae-
three feline hemoplasma species also have been described. mocanis is a coccoid organism that often forms long chains of
‘Ca. M. turicensis’ was first described in a cat from Switzer- organisms (Figure 41-2). The prevalence of this infection is par-
land that had severe intravascular hemolysis (turicensis pertains ticularly high in kennel-raised dogs, which are often infected sub-
to Turicum, the Latin name of Zurich).11 Infections with ‘Ca. M. clinically.18 In the southwestern United States, infection was also
turicensis’ have since been detected worldwide.8,12-14 ‘Ca. M. turi- prevalent among coyotes.19
censis’ has never been seen using light microscopic examination Three additional hemoplasma species have been detected in
of blood smears, and organism loads in infected cats are typically dogs. ‘Candidatus Mycoplasma haematoparvum’ is a small (0.3
low. Infection with ‘Ca. M. turicensis’ is slightly more prevalent in µm) coccoid organism that resembles ‘Ca. Mycoplasma haemo-
the cat population than infection with M. haemofelis. Most stud- minutum’ morphologically as well as genetically (Figure 41-3).
ies show a prevalence of 0.5% to 10% in sick cats that visit vet- ‘Ca. M. haemominutum’ has also been detected in several dogs
erinary hospitals. The pathogenic potential of this organism is not using PCR assays, and organisms that resemble ‘Ca. M. haema-
fully understood. Inoculation of an immunosuppressed cat with toparvum’ and ‘Ca. M. haemominutum’ have been detected in
‘Ca. M. turicensis’ resulted in severe anemia,11 but little or no ane- European wolves and bush dogs from Brazil.20 The ovine hemo-
mia occurs in immunocompetent cats after inoculation with ‘Ca. plasma Mycoplasma ovis was detected in the spleens of a small
M. turicensis.’ Cofactors, such as co-infection with other hemo- number of dogs from the southeastern United States, and the
plasmas or concurrent immunosuppression, may influence the bovine hemoplasma ‘Ca. Mycoplasma haemobos’ was detected
development of anemia in cats infected with ‘Ca. M. turicensis.’ in a dog from northern Australia.21,22 The clinical importance of
Infection of cats by hemoplasmas is strongly associ- these hemoplasma species in dogs remains unclear.
ated with male sex, nonpedigree status, and outdoor access The mode of transmission of hemoplasmas remains unclear.
(Box 41-1).8,12,15,16 Infection with ‘Ca. M. haemominutum’ is To some extent, fleas and other arthropod vectors may be capa-
more prevalent in older cats, presumably because the chance ble of transmitting feline hemoplasmas,23 but experimental evi-
dence for vector-borne transmission of feline hemoplasmas is
weak. Transmission of M. haemocanis by the brown dog tick,
BOX 41-1 Rhipicephalus sanguineus, has been demonstrated experimen-
tally, although this was before PCR assays were available to
Risk Factors Associated with Infection by Feline Hemotropic confirm infection.24 Geographic variation in the prevalence of
Mycoplasmas as Determined Using Species-Specific
Real-Time PCR Assays

Mycoplasma haemofelis
Younger age
Male sex
FIV seroreactivity
FeLV seroreactivity

‘Candidatus Mycoplasma haemominutum’


Older age
Male sex
FIV seroreactivity
Nonpedigree status
Outdoor access

‘Candidatus Mycoplasma turicensis’


Male sex

From Ettinger SJ, Feldman EC, eds. Textbook of Veterinary Internal FIGURE 41-2  Romanowsky-stained blood smear showing chains of epierythrocytic
Medicine, 7 ed, St. Louis, MO: Saunders; 2011. bacteria typical of Mycoplasma haemocanis. A Howell-Jolly body is also present (arrow).
392 SECTION 2  Bacterial Diseases

FIGURE 41-3  Blood smear from a dog showing ‘Candidatus Mycoplasma haema-
toparvum’ (small arrows) and a Howell-Jolly Body (large arrow).

hemoplasma infection in dogs and cats supports a role for arthro-


pod vectors in transmission. For example, in Europe, infection
with M. haemocanis is more prevalent in Mediterranean coun-
tries, which follows the distribution of Rh. sanguineus. Vertical
(e.g., transplacental) spread has also been hypothesized and has
been documented for bovine hemoplasmas. Biting has been sug-
gested as a means of transmission of feline hemoplasmas, and FIGURE 41-4  Electron micrographs showing epierythrocytic location of Mycoplasma
haemofelis.
the strong male sex predilection, recent history of cat bite absces-
sation in some cats, and association with FIV infection in some
studies supports this mode. Additionally, studies in Switzerland
found that subcutaneous inoculation of blood that contained erythrocyte life span have been noted in infected cats.27-29 The
‘Ca. M. turicensis’ resulted in transmission, whereas inoculation organisms can also “bridge” adjacent erythrocytes, which might
of saliva that contained ‘Ca. M. turicensis’ did not.25 This sug- promote splenic trapping and removal of red blood cells. In
gests that hemoplasma transmission by social contact (saliva via contrast, erythrocyte-bound antibody formation and significant
mutual grooming etc.) is less likely than transmission by aggres- reticulocytosis have not been documented in cats infected with
sive interaction (blood transmission during a cat bite incident). ‘Ca. M. haemominutum’ or ‘Ca. M. turicensis.’
Because infection can also be transmitted by ingestion of blood, Anemia can result in signs of lethargy, inappetence, pallor,
it may be that the biting cat (rather than the bitten cat) is most at and weakness. Some owners report that their cats eat dirt or
risk for acquisition of infection. Transmission of M. haemocanis litter or lick cement (pica). Weight loss and dehydration can
through ingestion of infected blood has also been described,26 so occur. Rapid development of anemia may result in neurologic
aggressive interactions between dogs may also have the poten- signs, vocalization, collapse, and death. In some infected cats,
tial to transmit hemoplasmas; however, this mode of transmis- cyclical changes in the hematocrit and numbers of infected
sion remains to be proven in field circumstances. Transmission erythrocytes occur, with sharp declines in the hematocrit corre-
can also occur following blood transfusion. lating with appearance of large numbers of organisms in blood
smears.30-32 The proportion of erythrocytes associated with vis-
Clinical Features ible organisms on blood smears can decline from 90% to less
than 1% in less than 3 hours.33,34 These dramatic fluctuations
Signs and Their Pathogenesis in organism numbers appear to result from rapid replication of
Because only M. haemofelis infection is associated with signifi- M. haemofelis, followed by rapid clearance from the blood as a
cant anemia in cats, only the pathogenesis of disease induced by result of the host’s immune response. Repeated antigenic varia-
this species is described here. After experimental infection, there tion in M. haemofelis may allow the organism to again prolifer-
is a delay of 2 to 34 days before the onset of clinical signs. Anemia ate in the face of this immune response, which contributes to
then occurs and persists for about 18 to 30 days (acute phase), ongoing fluctuations in organism numbers.
although its severity and chronicity vary considerably between In cats that survive the acute phase, the hematocrit then
infected cats. Anemia predominantly results from extravascu- returns to normal or near-normal (recovery phase), and organ-
lar hemolysis, and the onset of anemia is usually followed by isms can no longer be visualized in blood smears. It is possible
a strong regenerative response with reticulocytosis. The organ- that at least some of these recovered cats remain persistently
ism resides in an indentation on the erythrocyte surface (Figure infected, whereby the organism evades the host immune system,
41-4). Production of both cold and warm reactive erythrocyte- and that recrudescence of anemia may follow stress, pregnancy,
bound antibodies, increased osmotic fragility, and decreased intercurrent infection, or neoplasia.32,33 However, evidence is
CHAPTER 41  Hemoplasma Infections 393

accumulating that differences may exist in the ability of hemo-


plasmas to persist within the host, the carrier state being more
frequent for ‘Ca. M. haemominutum’ but less frequent for M.
haemofelis and ‘Ca. M. turicensis.’ In cats, splenectomy has a
variable effect on the course of hemoplasmosis. Recrudescence
of anemia and bacteremia has been documented in some chroni-
cally infected cats after splenectomy, although other studies sug-
gest that splenectomy increases the number of visible organisms
in blood smears without causing significant anemia.32,34 Infec-
tion of splenectomized cats with ‘Ca. M. haemominutum’ does
not appear to enhance the pathogenicity of this organism.35
Dogs that develop clinical hemoplasmosis have usually had a
history of splenectomy or, less commonly, concurrent immuno-
suppressive illness or drug treatment or co-infections with other
bloodborne pathogens such as Babesia spp. and Ehrlichia canis.
Clinical signs include weakness, lethargy, and pallor, and some
dogs are inappetent.
FIGURE 41-5  Mild icterus of the nictating membrane in an 8-year-old male neutered
Physical Examination Findings domestic shorthair with hemolytic anemia secondary to Mycoplasma haemofelis infection.
Physical examination findings in cats with acute M. haemofelis
infection include fever, weakness, mucosal pallor, tachypnea,
tachycardia, and weak or bounding femoral pulse quality. Other
physical examination abnormalities can include dehydration,
cardiac murmurs, splenomegaly, and occasionally mild icterus
(Figure 41-5). Moribund cats may be hypothermic.
Dogs with hemoplasmosis can show lethargy and mucosal
pallor. Fever is usually absent.
Cats infected with ‘Ca. M. haemominutum’ or ‘Ca. M. turi-
censis,’ and dogs infected with ‘Ca. M. haematoparvum’ gener-
ally appear healthy unless concurrent disease is present.

Diagnosis
Laboratory Abnormalities
Complete Blood Count
The most characteristic CBC abnormality in dogs and cats
infected with M. haemocanis and M. haemofelis, respectively, is
regenerative anemia, with macrocytosis, anisocytosis, reticulo-
cytosis, polychromasia, Howell-Jolly bodies, and, especially in
cats, sometimes marked normoblastemia. Manual reticulocyte
counts should be interpreted with caution in cats infected with
M. haemofelis, because both hemoplasma-infected erythrocytes
have the same appearance as reticulocytes in blood smears stained
with methylene blue. Autoagglutination may be noted in blood
smears. Nonregenerative anemia can be present when sufficient FIGURE 41-6  Ventrodorsal abdominal radiograph from an 8-year-old male neutered
time for a regenerative response has not yet elapsed, or as a result domestic shorthair cat with hemolytic anemia secondary to Mycoplasma haemofelis Infec-
of concurrent FeLV infection or any other disease that suppresses tion. There is marked splenomegaly (arrows).
the regenerative response. Concurrent occult hemoplasma infec-
tion should be considered in any anemic FeLV-positive cat, even
in the absence of reticulocytosis. Neutrophil counts may be nor- Diagnostic Imaging
mal, elevated, or low, and lymphopenia may be present. Throm- The most common radiographic finding in cats with hemoplas-
bocytopenia can also occur in affected dogs and cats, but often mosis is splenomegaly (Figure 41-6). The spleen may be absent
the platelet count is within the reference range. in affected dogs because of previous splenectomy.

Serum Biochemical Tests Microbiologic Tests


The serum chemistry profile in cats with hemoplasmosis may Assays available for diagnosis of hemoplasmosis in dogs and
reveal increased activities of ALT and AST as a result of hypoxia; cats are listed in Table 41-1.
metabolic acidosis; mild to moderate hyperbilirubinemia; and
prerenal azotemia. Hyperproteinemia may be seen in some cats. Cytologic Examination of Blood Smears
Cytologic detection of hemoplasmas has low sensitivity and
Urinalysis specificity, especially for diagnosis of feline infectious anemia.
The urinalysis is usually unremarkable; bilirubinuria may be M. haemofelis is visible less than 50% of the time in acutely
present in hyperbilirubinemic cats. infected cats, because organisms can disappear for days before
394 SECTION 2  Bacterial Diseases

TABLE 41-1
Assays Available for Diagnosis of Hemoplasmosis in Dogs and Cats
Assay Specimen Type Target Performance
Cytologic Blood smears Hemoplasmas Poor sensitivity, especially in affected cats. Organisms are seen on
examination blood smears in fewer than 50% of sick cats. False positives can
occur when stain precipitate, drying artifact, basophilic stippling,
and Howell-Jolly bodies are misinterpreted as hemoplasmas.
PCR Whole blood, blood Hemoplasma DNA Sensitivity and specificity can vary depending on assay design.
smears Results must be interpreted in light of the species detected and
the clinical findings. Mycoplasma haemofelis and Mycoplasma
haemocanis are most likely to be associated with anemia.

reappearing on blood smears over the course of infection. be tested but are less sensitive than liquid whole blood speci-
Because organisms detach from erythrocytes within hours mens.38 PCR assays designed to detect M. haemofelis generally
of blood collection, fresh blood smears should be submitted. also detect M. haemocanis because of the high 16S rRNA gene
False-positive diagnoses occur when stain artifacts such as pre- sequence homology between these species, but a separate assay
cipitate are confused with organisms; thus, careful staining with is required for detection of ‘Ca. M. haematoparvum.’
a properly prepared, uncontaminated Romanowsky-type stain
solution is essential. Organisms also need to be distinguished Serologic Diagnosis
from basophilic stippling and Howell-Jolly bodies. ‘Ca. M. hae- The development of serologic tests for diagnosis of hemoplas-
mominutum’ is small and is not usually visible in chronically mosis has been hampered by the inability to culture hemoplas-
infected cats. ‘Ca. M. turicensis’ has never been seen on blood mas in the laboratory. However, the application of molecular
smears with light microscopy. Hemoplasmas are visible in many techniques has led to the identification of antigens for serologic
clinically affected dogs, and because M. haemocanis tends to assay development. Specifically, an ELISA that detects antibod-
form chains, it is more readily differentiated from stain artifacts ies to an immunodominant protein of M. haemofelis, DnaK,
and erythrocyte morphologic changes than is M. haemofelis. has been developed. Cats produce antibodies to this antigen
However, hemoplasmas are not usually visible in chronically after infection,39,40 and in a study that examined the immune
infected dogs or in dogs infected with ‘Ca. M. haematoparvum.’ response to ‘Ca. M. turicensis’ infection, antibody titers to
DnaK declined with antimicrobial drug treatment.39 Serologic
Culture cross-reactivity occurs to this antigen for all three feline hemo-
Attempts to isolate and grow feline hemotropic mycoplasmas in plasma species40; therefore, the assay does not discriminate
the laboratory have been unsuccessful, so blood culture cannot between infection with M. haemofelis and other hemoplasmas,
be used for diagnosis of hemoplasmosis. but determination of acute and convalescent titers may be help-
ful to distinguish acute from chronic infection. Currently this
Molecular Diagnosis Using the Polymerase Chain Reaction assay is available only on a research basis.
PCR assays are currently the tests of choice for diagnosis of
hemoplasmosis in dogs and cats. Numerous assays have been Pathologic Findings
described to date, which are generally based on detection of Generalized pallor, splenomegaly, and, in some cats, icterus are
the 16S rRNA gene. These are significantly more sensitive than the main gross necropsy finding in cats that die or are eutha-
blood smear evaluation, although they occasionally do not nized as a result of hemoplasmosis. Histopathologic findings
detect infection in apparently healthy carrier cats when organism include extramedullary hematopoiesis, follicular hyperplasia,
numbers in the blood are below the detection limit of the PCR and erythrophagocytosis within the spleen.
assay.6,30,36,37 Both conventional and real-time PCR assays have
been described. Some conventional PCR assays do not differ- Treatment and Prognosis
entiate between M. haemofelis and ‘Ca. M. turicensis,’ because
the primers used in the assay generate the same-sized PCR prod- Treatment is indicated for cats and dogs with clinical signs
uct when either or both of these species are present. Real-time and laboratory abnormalities consistent with hemoplasmosis.
PCR assays are usually species specific. Because the pathogenic Although controversial, treatment could also be considered
potential of each hemoplasma species differs, the laboratory for apparently healthy cats that test positive for M. haemofelis
should be consulted to determine the species specificity of the in view of the potential for recrudescence of disease with this
assay(s) offered. Alternative causes of anemia should always be hemoplasma species. It may be difficult or impossible to eradi-
considered, especially in cats that test positive for ‘Ca. M. hae- cate infection with some hemoplasma species or strains, espe-
mominutum’ or ‘Ca. M. turicensis,’ because infection with these cially ‘Ca. M. haemominutum.’ Indeed, no antibiotic treatment
organisms is not commonly associated with anemia. Indeed, regime consistently eliminates hemoplasma infection in cats.
all PCR results should be interpreted in conjunction with the The treatment of choice for hemoplasmosis is doxycycline
patient’s clinical signs, the degree and nature of the anemia pres- for a minimum of 2 weeks (see Chapter 8 for information on
ent, and any concurrent signs or diseases present that could be doxycycline) (Table 41-2); some have suggested using longer
contributing to the clinical signs. Dried blood smears can also courses of treatment (6 to 8 weeks) to ensure that infection is
CHAPTER 41  Hemoplasma Infections 395

Immunity and Vaccination


TABLE 41-2
Suggested Drug Dosages for Treatment of Hemoplasmosis Little information is available in regard to the immune response
to hemoplasma infection, and no vaccines are available. Because
in Cats
co-infections with multiple hemoplasma species occur, infection
with one species does not appear to protect against infection
Dose Interval
with other species.
Drug (mg/kg) Route (hours) Duration
Doxycycline 10 PO 24 2 weeks Prevention
Marbofloxacin 2.75 to 5.5 PO 24
Pradofloxacin* 5 to 10 PO 24 Inadvertent transmission of hemoplasmas by transfusion of
blood from carrier cats has been documented, and so all blood
Enrofloxacin† 5 PO 24 donor dogs and cats should be tested for hemoplasmas with
*Dose listed is for the oral suspension for cats. PCR assays. In the future, screening with serologic assays may
†Only when other alternatives are not available because of potential for also be used to identify potential donor animals. Whenever pos-
irreversible blindness when enrofloxacin is used in cats. sible, cats and dogs that are PCR negative for any of the known
hemoplasma species should be excluded as donors. Given the
high prevalence of subclinical ‘Ca. M. haemominutum’ infec-
eliminated. Real-time PCR could be used to monitor response tion among cats, in some circumstances it may not be possible
to antibiotic treatment if quantitative information is avail- to find a donor that is ‘Ca. M. haemominutum’ negative. At this
able. Fluoroquinolones are an alternative; marbofloxacin and time, it is unclear whether transfusion of blood from cats that are
pradofloxacin,41 when available, are both efficacious. Enro- infected with ‘Ca. M. haemominutum’ is associated with adverse
floxacin is also usually effective,42 but diffuse retinal degenera- outcome, but it remains a possibility. Keeping cats indoors is
tion and acute blindness have been reported following the use also likely to prevent infection, as outdoor access has been iden-
of this fluoroquinolone and, although this is rare, caution must tified as a risk factor. Control of fleas and ticks is recommended.
be exercised with its use in cats. The response of the different
hemoplasma species, and indeed different strains of the same Public Health Aspects
species, to antibiotics varies. Most successful studies that have
demonstrated treatment efficacy have evaluated the response Organisms that resembled hemoplasmas were identified in
of M. haemofelis to treatment. Affected dogs and cats may human blood smears decades ago. Molecular techniques that
also require supportive treatment with IV crystalloids or blood were originally designed for detection of canine and feline hemo-
products. Clinical improvement usually occurs within 2 to plasmas have since been applied to human blood, and a variety
3 days. The use of immunosuppressive doses of glucocorticoids of hemoplasma species have now been identified. In addition,
to suppress the associated immune-mediated hemolytic process is application of these methods to archived human blood smears
controversial, given that glucocorticoids can cause reactivation of believed to contain hemoplasmas based on cytological evaluation
latent infection, but may be necessary in cats that fail to respond yielded negative results, suggesting the possibility of historical
to antimicrobial therapy alone, or when the diagnosis is uncer- misdiagnosis.45 Infection with a novel hemoplasma, designated
tain, especially if erythrocyte-bound antibodies are identified. ‘Ca. Mycoplasma haemohominis,’ has been identified in a per-
Doxycycline is the drug of choice for treatment of M. hae- son with hemolytic anemia.46 M. haemofelis DNA was identi-
mocanis infection. Treatment of a splenectomized dog with fied in a person from Brazil who was co-infected with HIV and
oxytetracycline for approximately 1 month was associated Bartonella henselae.47 In addition, DNA of Mycoplasma ovis
with clinical improvement, but infection persisted as deter- was detected in a Texas veterinarian who was co-infected with
mined with quantitative PCR, and relapse occurred when B. henselae.48 Mycoplasma suis DNA has been found in farm
treatment was withdrawn.43 Infection also persisted after workers from China.49 The identification of the DNA of animal
treatment with enrofloxacin, despite eventual recovery from hemoplasmas in humans should be viewed cautiously because
anemia. In contrast, in M. haemocanis–infected dogs with only a small portion or portions of the hemoplasma genome
intact spleens, doxycycline treatment for 4 weeks appeared to was detected in each of these case reports. M. haemocanis and
result in resolution of infection as determined by PCR assay, M. haemofelis have identical 16S rRNA gene sequences, but
whereas ‘Ca. M. haematoparvum’–infected dogs remained different host tropisms, and so the organisms detected in these
PCR positive.19 A splenectomized dog was apparently cured humans may not necessarily be the same as those that infect
of M. haemocanis infection after 12 weeks of doxycycline domestic animal species. Until further information is available,
treatment.44 veterinarians should handle blood from animals with caution.
396 SECTION 2  Bacterial Diseases

CASE EXAMPLE Hb = 2.8 g/dL, O2 = 59.5%, potassium = 4.0 mmol/L, sodium =


144 mmol/L, ionized calcium = 1.2 mmol/L, glucose = 157 mg/
dL, lactate = 8.4 mmol/L, base excess = −12.2 mmol/L.
Signalment: “Shadow,” an 8-year-old male neutered Imaging Findings:
domestic shorthair from Fairfield, CA Thoracic Radiographs: No clinically significant abnormalities
History: Shadow was brought to the University of California, were detected.
Davis, Veterinary Medical Teaching Hospital emergency Abdominal Radiographs: The spleen was markedly enlarged
service for treatment of severe anemia. The owners reported (See Figure 41-6). The liver was mildly enlarged. Small
that he had disappeared 5 days previously. The evening that mineral densities were present within ingesta and feces in
he was brought to the emergency service, he was found the stomach and colon, respectively.
splayed across a fence and was unwilling to move, and Treatment: The cat was placed on a heating pad and
vocalizing and salivating profusely. He was immediately transfused with 1 unit of packed RBC, and supplemental
taken to a local emergency clinic where he was found to oxygen was administered, after which heart rate increased
be obtunded, pale, and severely dehydrated. His pulse was to 200 beats/min and respiratory rate decreased to 40
180 beats/min, respiratory rate was 60 breaths/min, and breaths/min. Rectal temperature increased to 103.7°F
temperature was 94.0°F (34.4°C). A systolic blood pressure (39.8°C) during the transfusion, and so dexamethasone
was 80 mm Hg. A PCV was 12%. Shadow’s condition was was administered because of the concern for a transfusion
stabilized after treatment with supplemental oxygen, active reaction (0.2 mg/kg IV once). The cat was also treated with
warming, and IV crystalloid fluids (Normosol-R, 50 mL over enrofloxacin (5 mg/kg IV q24h) (see comments section later)
30 minutes, then 25 mL/hr). He tested negative for FIV for suspected hemoplasmosis. The results of laboratory
antibody and FeLV antigen with an in-practice ELISA assay. testing after transfer to the internal medicine service the day
Shadow was an indoor-outdoor cat that was acquired as a after admission are shown here.
stray kitten from the owner’s backyard. There had been no CBC (Post Transfusion and Antimicrobial Treatment):
previous history of illness. HCT 12.7% (30%-50%)
Physical Examination: MCV 76.5 fL (42-53 fL)
Body Weight: 6 kg MCHC 26.8 g/dL (30-33.5 g/dL)
General: Obtunded, laterally recumbent. T = 97.8°F (36.6°C), HR Reticulocytes 155,600 cells/µL (7000-60,000 cells/µL)
= 150 beats/min, RR = 60 breaths/minute, purring, pale and Nucleated RBC 39/100 WBC (0/100 WBC)
tacky mucous membranes with no detectable capillary refill, Corrected WBC 9800 cells/µL (4500-14,000 cells/µL)
weak pulses. Neutrophils 8134 cells/µL (2000-9000 cells/µL)
Integument: No ectoparasites were noted. The pinnae and Band neutrophils 98 cells/µL
ventral abdominal skin were icteric. Lymphocytes 784 cells/µL (1000-7000 cells/µL)
Eyes, Ears, Nose, and Throat: Moderate dental calculus was Monocytes 784 cells/µL (50-600 cells/µL)
present. Pallor and mild icterus of the mucous membranes, Eosinophils 0 cells/µL (150-1100 cells/µL)
nictitans, and sclera was noted (see Figure 41-5). Basophils 0 cells/µL (0-50 cells/µL)
Musculoskeletal: Body condition score was 7/9. Platelets 142,000/µL (180,000-500,000 platelets/µL).
Cardiovascular: Weak pulses. No other clinically significant Cytologic examination revealed marked anisocytosis, marked
abnormalities were detected. No murmurs were detected polychromasia, few Howell-Jolly bodies, and many mac-
but the heart sounds were difficult to auscultate because of rocytes and microcytes. No hemoplasmas were identified.
persistent purring. Serum Chemistry Profile:
Respiratory: Tachypneic; auscultation was limited because of Sodium 143 mmol/L (151-158 mmol/L)
purring. Potassium 5.2 mmol/L (3.6-4.9 mmol/L)
Gastrointestinal and Genitourinary: Nonpainful abdomen. Chloride 107 mmol/L (117-126 mmol/L)
Moderate cranial organomegaly. Urinary bladder not Bicarbonate 11 mmol/L (15-21 mmol/L)
palpable. Phosphorus 6.6 mg/dL (3.2-6.3 mg/dL)
Neurologic: A full neurologic examination was not performed Calcium 9.6 mg/dL (9.0-10.9 mg/dL)
but no abnormalities were detected on examination of BUN 47 mg/dL (18-33 mg/dL)
cranial nerve functions. Creatinine 1.2 mg/dL (1.1-2.2 mg/dL)
Lymph Nodes: No clinically significant abnormalities Glucose 98 mg/dL (63-118 mg/dL)
detected. Total protein 7.2 g/dL (6.6-8.4 g/dL)
Laboratory Findings: Albumin 3.0 g/dL (2.2-4.6 g/dL)
PCV/TPP: 14%/7.3 g/dL, icteric plasma. Globulin 4.2 g/dL (2.8-5.4 g/dL)
Saline Slide Agglutination Test: Negative for ALT 166 U/L (27-101 U/L)
autoagglutination. AST 201 U/L (17-58 U/L)
Blood Smear Examination: Marked polychromasia, ALP 24 U/L (14-71 U/L)
normoblastosis, and mild anisocytosis were present. GGT 0 U/L (0-4 U/L)
Coccoid bodies that resembled hemoplasmas were present Cholesterol 75 mg/dL (89-258 mg/dL)
in association with erythrocytes (see Figure 41-1). Total bilirubin 6.1 mg/dL (0-0.2 mg/dL)
Electrolyte/Acid-Base Panel (Venous Blood): HCO3 = 14 Magnesium 3.4 mg/dL (1.5-2.5 mg/dL)
mmol/L, pH = 7.365, pCO2 = 21.9 mm Hg, pO2 = 35.4 mm Hg, Creatine kinase 446 U/L (73-260 U/L).
CHAPTER 41  Hemoplasma Infections 397

Urinalysis: USG 1.040, pH 6, 75 mg/dL protein, no glucose, Comments: This represents a typical case of hemoplasmosis in a
5 mg/dL ketones, 6 mg/dL bilirubin, 250 erythrocytes/µL male neutered cat with access to the outdoors. Specific testing
hemoprotein, 0-1 RBC/HPF, 0 WBC/HPF, rare transitional for hemoplasmas with PCR was performed on blood smears
epithelial cells. because treatment with packed RBC and antibiotics had been
Microbiologic Testing: Real-time PCR for Mycoplasma initiated immediately on admission and whole blood was
haemofelis, ‘Candidatus Mycoplasma haemominutum,’ and not saved before treatment was instituted. Although PCR on
‘Candidatus Mycoplasma turicensis’ (blood smear): Positive blood smears has lower sensitivity than when whole liquid
for M. haemofelis DNA blood is used, in this case sufficient organisms were present
Diagnosis: Feline infectious anemia (Mycoplasma haemofelis to generate a positive result, which confirmed the diagnosis
infection) of hemoplasmosis. Because the cat was hypovolemic on
Additional Treatment and Outcome: Treatment was admission, parenteral antimicrobial treatment was chosen.
changed to doxycycline (10 mg/kg followed by 5 mL of Subsequently, treatment with oral doxycycline was initiated.
water PO q24h for 3 weeks). Treatment with IV crystalloid Although continued treatment with oral enrofloxacin may
fluids was continued and oxygen supplementation was have been a suitable alternative to doxycycline, retinal
discontinued. Two days after admission, the PCV was 16% degeneration was a possible adverse effect of continued
and the cat became aggressive and began eating. The cat treatment with enrofloxacin. Retesting for retroviruses was
was discharged the following day with a PCV of 18%, and indicated 2 months after the first test was performed at the
a recheck at the local veterinary clinic that included a CBC local veterinary clinic, as the cat may have been exposed to
was recommended 1 week later. The owner subsequently retroviruses but not yet have developed positive test results
elected to house Shadow indoors. (see Chapters 21 and 22).

SUGGESTED READINGS 11. Willi B, Boretti FS, Cattori V, et al. Identification, molecular char-
acterization, and experimental transmission of a new hemoplasma
Dowers KL, Tasker S, Radecki SV, et al. Use of pradofloxacin to treat isolate from a cat with hemolytic anemia in Switzerland. J Clin
experimentally induced Mycoplasma haemofelis infection in cats. Am Microbiol. 2005;43:2581-2585.
J Vet Res. 2009;70:105-111. 12. Willi B, Tasker S, Boretti FS, et al. Phylogenetic analysis of ‘Candi-
Tasker S, Helps CR, Day MJ, et al. Use of real-time PCR to detect and datus Mycoplasma turicensis’ isolates from pet cats in the United
quantify Mycoplasma haemofelis and “Candidatus Mycoplasma hae- Kingdom, Australia, and South Africa, with analysis of risk factors
mominutum” DNA. J Clin Microbiol. 2003;41:439-441. for infection. J Clin Microbiol. 2006;44:4430-4435.
13. Peters IR, Helps CR, Willi B, et al. The prevalence of three species
REFERENCES of feline haemoplasmas in samples submitted to a diagnostics ser-
1. Clark R. Eperythrozoon felis (sp. nov.) in a domestic cat. J S Afr vice as determined by three novel real-time duplex PCR assays. Vet
Vet Med Assoc. 1942;13:15-16. Microbiol. 2008;126:142-150.
2. Kikuth W. Über einen neuen Anämeerreger: Bartonella canis nov. 14. Tanahara M, Miyamoto S, Nishio T, et  al. An epidemiological
spec. Klin Wochenschr. 1928;7:1729-1730. survey of feline hemoplasma infection in Japan. J Vet Med Sci.
3. Barker EN, Darby AC, Helps CR, et al. Genome sequence for ‘Can- 2010;72:1575-1581.
didatus Mycoplasma haemominutum,’ a low-pathogenicity hemo- 15. Sykes JE, Terry JC, Lindsay LL, et al. Prevalences of various hemo-
plasma species. J Bacteriol. 2012;194:905-906. plasma species among cats in the United States with possible hemo-
4. Barker EN, Helps CR, Peters IR, et al. Complete genome sequence plasmosis. J Am Vet Med Assoc. 2008;232:372-379.
of Mycoplasma haemofelis, a hemotropic mycoplasma. J Bacteriol. 16. Willi B, Boretti FS, Baumgartner C, et  al. Prevalence, risk fac-
2011;193:2060-2061. tor analysis, and follow-up of infections caused by three feline
5. Santos AP, Guimaraes AM, do Nascimento NC, et al. Genome of hemoplasma species in cats in Switzerland. J Clin Microbiol.
Mycoplasma haemofelis, unraveling its strategies for survival and 2006;44:961-969.
persistence. Vet Res. 2011;42:102. 17. Nascimento NC, Santos AP, Guimaraes AM, et  al. Mycoplasma
6. Jensen WA, Lappin MR, Kamkar S, et al. Use of a polymerase chain haemocanis str. Illinois chromosome, complete genome. In:
reaction assay to detect and differentiate two strains of Haemobarton- National Center for Biotechnology Information, January 11, 2012
ella felis in naturally infected cats. Am J Vet Res. 2001;62:604-608. ed. Bethesda, MD: National Institutes of Health; 2012.
7. Tasker S, Binns SH, Day MJ, et al. Use of a PCR assay to assess the 18. Kemming GI, Messick JB, Enders G, et al. Mycoplasma haemocanis
prevalence and risk factors for Mycoplasma haemofelis and ‘Candi- infection—a kennel disease? Comp Med. 2004;54:404-409.
datus Mycoplasma haemominutum’ in cats in the United Kingdom. 19. Sykes JE, unpublished data, 2012.
Vet Rec. 2003;152:193-198. 20. André MR, Adania CH, Allegretti SM, et al. Hemoplasmas in wild
8. Sykes JE, Drazenovich NL, Ball LM, et al. Use of conventional and canids and felids in Brazil. J Zoo Wildl Med. 2011;42:342-347.
real-time polymerase chain reaction to determine the epidemiol- 21. Varanat M, Maggi RG, Linder KE, et al. Molecular prevalence of
ogy of hemoplasma infections in anemic and nonanemic cats. J Vet Bartonella, Babesia, and hemotropic Mycoplasma sp. in dogs with
Intern Med. 2007;21:685-693. splenic disease. J Vet Intern Med. 2011;25:1284-1291.
9. George JW, Rideout BA, Griffey SM, et  al. Effect of preexisting 22. Hii SF, Kopp SR, Thompson MF, et al. Canine vector-borne dis-
FeLV infection or FeLV and feline immunodeficiency virus coinfec- ease pathogens in dogs from south-east Queensland and north-east
tion on pathogenicity of the small variant of Haemobartonella felis Northern Territory. Aust Vet J. 2012;90(4):130-135.
in cats. Am J Vet Res. 2002;63:1172-1178. 23. Woods JE, Brewer MM, Hawley JR, et  al. Evaluation of experi-
10. Reynolds CA, Lappin MR. ‘Candidatus Mycoplasma haemominu- mental transmission of ‘Candidatus Mycoplasma haemominutum’
tum’ infections in 21 client-owned cats. J Am Anim Hosp Assoc. and Mycoplasma haemofelis by Ctenocephalides felis to cats. Am J
2007;43:249-257. Vet Res. 2005;66:1008-1012.
398 SECTION 2  Bacterial Diseases

24. Seneviratna P, Weerasinghe, Ariyadasa S. Transmission of Haemo- 38. Sykes JE, Owens SD, Terry JC, et al. Use of dried blood smears for
bartonella canis by the dog tick, Rhipicephalus sanguineus. Res Vet detection of feline hemoplasmas using real-time polymerase chain
Sci. 1973;14:112-114. reaction. J Vet Diagn Invest. 2008;20:616-620.
25. Museux K, Boretti FS, Willi B, et al. In vivo transmission studies of 39. Novacco M, Wolf-Jackel G, Riond B, et  al. Humoral immune
‘Candidatus Mycoplasma turicensis’ in the domestic cat. Vet Res. response to a recombinant hemoplasma antigen in experimental
2009;40:45. ‘Candidatus Mycoplasma turicensis’ infection. Vet Microbiol.
26. Lumb WV. Haemobartonellosis in the dog. In: 8th Gaines Veteri- 2012;157:464-470.
nary Symposium 1958;15–16. 40. Barker EN, Helps CR, Heesom KJ, et  al. Detection of humoral
27. Maede Y. Studies on feline haemobartonellosis. IV. Lifespan of response using a recombinant heat shock protein 70, DnaK, of
erythrocytes of cats infected with Haemobartonella felis. Nihon Mycoplasma haemofelis in experimentally and naturally hemo-
Juigaku Zasshi. 1975;37:269-272. plasma-infected cats. Clin Vaccine Immunol. 2010;17:1926-1932.
28. Maede Y, Hata R. Studies on feline haemobartonellosis. II. The 41. Dowers KL, Tasker S, Radecki SV, et al. Use of pradofloxacin to
mechanism of anemia produced by infection with Haemobarton- treat experimentally induced Mycoplasma haemofelis infection in
ella felis. Nihon Juigaku Zasshi. 1975;37:49-54. cats. Am J Vet Res. 2009;70:105-111.
29. Zulty JC, Kociba GJ. Cold agglutinins in cats with haemobartonel- 42. Tasker S, Helps CR, Day MJ, et al. Use of a Taqman PCR to deter-
losis. J Am Vet Med Assoc. 1990;196:907-910. mine the response of Mycoplasma haemofelis infection to antibiotic
30. Foley JE, Harrus S, Poland A, et al. Molecular, clinical, and patho- treatment. J Microbiol Methods. 2004;56:63-71.
logic comparison of two distinct strains of Haemobartonella felis in 43. Hulme-Moir KL, Barker EN, Stonelake A, et  al. Use of real-
domestic cats. Am J Vet Res. 1998;59:1581-1588. time quantitative polymerase chain reaction to monitor antibiotic
31. Tasker S, Helps CR, Day MJ, et al. Use of real-time PCR to detect therapy in a dog with naturally acquired Mycoplasma haemocanis
and quantify Mycoplasma haemofelis and ‘Candidatus Myco- infection. J Vet Diagn Invest. 2010;22:582-587.
plasma haemominutum’ DNA. J Clin Microbiol. 2003;41:439-441. 44. Pitorri F, Dell’Orco M, Carmichael N, et al. Use of real-time quan-
32. Harvey DG, Gaskin JM. Feline haemobartonellosis: attempts to titative PCR to document the successful treatment of Mycoplasma
induce relapses of clinical disease in chronically infected cats. J Am haemocanis infection with doxycycline in a dog. Vet Clin Pathol.
Anim Hosp Assoc. 1978;14:453. 2012;41:493-496.
33. Harvey JW, Gaskin JM. Experimental feline haemobartonellosis. 45. Tasker S, Peters IR, Mumford AD, et  al. Investigation of human
J Am Anim Hosp Assoc. 1977;13:28. haemotropic Mycoplasma infections using a novel generic haemo-
34. Alleman AR, Pate MG, Harvey JW, et  al. Western immunob- plasma qPCR assay on blood samples and blood smears. J Med
lot analysis of the antigens of Haemobartonella felis with sera Microbiol. 2010;59:1285-1292.
from experimentally infected cats. J Clin Microbiol. 1999;37: 46. Steer JA, Tasker S, Barker EN, et  al. A novel hemotropic Myco-
1474-1479. plasma (hemoplasma) in a patient with hemolytic anemia and pyrexia.
35. Sykes JE, Henn JB, Kasten RW, et  al. Bartonella henselae infec- Clin Infect Dis. 2011;53:e147-151.
tion in splenectomized domestic cats previously infected with 47. dos Santos AP, dos Santos RP, Biondo AW, et  al. Hemoplasma
hemotropic Mycoplasma species. Vet Immunol Immunopathol. infection in HIV-positive patient. Brazil. Emerg Infect Dis.
2007;116:104-108. 2008;14:1922-1924.
36. Berent LM, Messick JB, Cooper SK. Detection of Haemobarton- 48. Sykes JE, Lindsay LL, Maggi RG, et al. Human coinfection with Bar-
ella felis in cats with experimentally induced acute and chronic tonella henselae and two hemotropic mycoplasma variants resem-
infections, using a polymerase chain reaction assay. Am J Vet Res. bling Mycoplasma ovis. J Clin Microbiol. 2010;48:3782-3785.
1998;59:1215-1220. 49. Yuan CL, Liang AB, Yao CB, et  al. Prevalence of Mycoplasma
37. Messick JB, Berent LM, Cooper SK. Development and evaluation suis (Eperythrozoon suis) infection in swine and swine-farm
of a PCR-based assay for detection of Haemobartonella felis in workers in Shanghai, China. Am J Vet Res. 2009;70:890-894.
cats and differentiation of H. felis from related bacteria by restric-
tion fragment length polymorphism analysis. J Clin Microbiol.
1998;36:462-466.
CHAPTER 42

Actinomycosis
Jane E. Sykes

Actinomycosis in outdoor dogs is often related to exposure to


Overview of Actinomycosis penetrating plant foreign bodies such as grass awns. In cats,
First Described: Actinomycosis was first described in 1877 (in actinomycosis usually follows bite wounds and so is more com-
cattle) (Otto Bollinger, Germany)1 monly diagnosed in males.18,19 In the author’s hospital, two-
thirds (11 of 17) cats with actinomycosis are male. However,
Causes: Actinomyces spp. and Arcanobacterium spp. (order
underlying retrovirus infection is uncommonly present and does
Actinomycetales, family Actinomycetaceae)
not appear to predispose to disease.
Geographic Distribution: Worldwide, but especially regions Because Actinomyces species are difficult to culture and are
where plant awns are prevalent susceptible to antimicrobial drugs that are often used empirically,
Mode of Transmission: Opportunistic invasion of commensal the prevalence of actinomycosis in dogs and cats is probably
bacteria following disruption of cutaneous or mucosal underestimated.
barriers
Clinical Features
Major Clinical Signs: Subcutaneous masses and draining
skin lesions (cervicofacial and cutaneous-subcutaneous Signs and Their Pathogenesis
disease); pulmonary nodules, masses, and/or effusion
After ingestion or inhalation, plant awns become contaminated
with cough and tachypnea (thoracic disease); abdominal
with Actinomyces spp. and other bacteria from the oropharynx,
effusion or masses (abdominal disease); thoracolumbar
and then migrate to various sites and act as a nidus of infection.
pain or pelvic limb paresis/paralysis (retroperitoneal dis-
Alternatively, organisms can be introduced into tissues at the
ease); rarely neurologic signs due to meningitis or brain
time of a bite wound injury. The latter is the most common route
abscesses (central nervous system disease).
of infection in cats, and can manifest as pyothorax, peritonitis, or
Differential Diagnoses: Mycobacterial infections, streptomyco- cellulitis.18,19 Other co-infecting aerobic and anaerobic bacteria
sis, nocardiosis, bartonellosis, fungal infections, neoplasia
Human Health Significance: Direct transmission of disease from
animals to humans does not occur, but humans have devel- BOX 42-1
oped actinomycosis after bites from healthy dogs or cats.
Actinomyces and Arcanobacterium Species Isolated from
Dogs and Cats with Actinomycosis
Etiology and Epidemiology
Dogs
Actinomycosis is caused by anaerobic or microaerophilic, Actinomyces viscosus2-4
filamentous, gram-positive bacteria that belong to the genus Actinomyces bowdenii5
Actinomyces (and to a lesser extent, the related genus Arcano- Actinomyces hordeovulneris6
bacterium). These organisms are normal inhabitants of mucous Actinomyces canis7
membranes, especially of the oropharynx, but also the genital Actinomyces odontolyticus8
and gastrointestinal tracts, and cause opportunistic infections. Actinomyces catuli9
Together with other oral commensal bacteria, actinomycetes Actinomyces turicensis10
colonize the periodontal mucosal surfaces and adhere to the Actinomyces hyovaginalis11
tooth surface to form plaque. Numerous Actinomyces species Actinomyces urogenitalis11
have been cultured from the mucous membranes, dental plaque, Arcanobacterium pyogenes12
and saliva of healthy dogs and cats. Organisms isolated from
ill dogs and cats with actinomycosis are shown in Box 42-1.2-15 Cats
Usually, actinomycosis develops when Actinomyces spp. Actinomyces viscosus13,14
are inoculated into tissues along with other bacteria, often as Actinomyces bowdenii5
a result of a deeply penetrating wound or foreign body migra- Actinomyces hordeovulneris11
tion. Young adult to middle-age large-breed dogs that have out- Actinomyces meyeri15
door access are often affected, especially retriever and hunting Actinomyces odontolyticus14
breeds.16,17 There is no clear sex predisposition, and the median Arcanobacterium pyogenes12
age of dogs with actinomycosis is approximately 5 years.18

399
400 SECTION 2  Bacterial Diseases

from the oral cavity or intestinal tract (“companion microbes”) pyogranulomatous inflammation (Figure 42-1). Large aggre-
undermine normal host defenses and reduce oxygen tension, gations of organisms form “sulfur granules,” which are tan to
which allows Actinomyces spp. to persist. Actinomyces spp. that yellow colonies of actinomycetes, which can be microscopic or
possess fimbriae can bind to specific cell surface receptors on visible grossly. Proteolytic enzymes from the associated bacteria,
other bacteria, especially streptococci. This bacterial co-aggrega- macrophages, and degranulated neutrophils destroy connective
tion inhibits the ability of neutrophils to phagocytize the organ- tissue, which facilitates extension of the disease through normal
isms.20 Dense colonies of Actinomyces spp. form, and these, tissue planes. Less often, organisms spread hematogenously to
together with other associated bacteria, are surrounded by neu- distant sites. In some cases, the inflammatory reaction is accom-
trophils, macrophages, and plasma cells, with slowly progressive panied by mass formation and extensive fibrosis. The center
of the lesions can eventually suppurate and soften, or draining
tracts may develop. The tracts can close and reappear over weeks
to months as infection gradually spreads through tissues.
The most common clinical forms of actinomycosis in cats
and dogs involve the cervicofacial region, thorax, abdomen,
and subcutaneous tissue, but central nervous system (CNS)
infections (meningitis and meningoencephalitis) and ocular
infections (keratitis and endophthalmitis) can also occur.21,22
Cervicofacial actinomycosis can follow bite wounds, perfora-
tion of the oropharynx by a foreign body, or chronic periodon-
tal disease. The mandibular, submandibular, and ventral or
lateral cervical areas are most frequently affected, but infections
that involve the face, retrobulbar space, and temporal area also
occur (Figure 42-2, A and B). Cutaneous-subcutaneous actino-
mycosis typically involves the lateral thoracic wall, flank region,
and occasionally the limbs. Lesions on the thoracic or abdomi-
nal walls may represent extensions of thoracic, abdominal, or
retroperitoneal actinomycosis.
Thoracic actinomycosis may be limited to the lung paren-
FIGURE 42-1  Abdominal fluid cytology from a 3-year-old male neutered domestic
longhair cat with a 1-week history of progressive lethargy and anorexia, ascites, hyper- chyma but can involve other thoracic structures, such as the
bilirubinemia, and hypoproteinemia. Abdominal fluid analysis revealed a total protein of mediastinum, pleura, pericardium, and thoracic wall. It usually
2.2 g/dL, and 75,500 nucleated cells/µL, with 92% neutrophils and 8% large mononuclear follows aspiration of oropharyngeal material, often together
cells. High numbers of moderately degenerative neutrophils were present with low num- with a contaminated grass awn. Alternative routes of thoracic
bers of macrophages. Moderate numbers of filamentous beaded bacteria were present infection include mediastinal involvement after esophageal
both in the background and within phagocytes (arrows). Actinomyces spp. and Fusobacte- perforation or direct extension of subcutaneous or abdomi-
rium nucleatum were isolated from the peritoneal fluid. Necropsy revealed severe, chronic nal disease. Clinical signs include cough and less commonly
peritonitis with intralesional filamentous bacteria. hemoptysis, tachypnea, respiratory distress, and subcutaneous

A B
FIGURE 42-2  Cervicofacial actinomycosis in an 8-year-old female spayed Labrador retriever with slowly progressive, left-sided facial swelling. Skull radiograph (A) and computed
tomographic scan (B). There was severe, regional soft tissue swelling over the left side of the face, which involved the left masseter muscle and zygomatic salivary glands and extended into
the retrobulbar space, with secondary exophthalmos. Multiple premolars and molars of the left maxillary arcade were missing. Biopsy of the lesion revealed severe, diffuse, pyogranulo-
matous cellulitis with intralesional gram-positive, acid-fast negative bacilli. Culture yielded Actinomyces hordeovulneris (based on 16S rRNA gene sequencing), Pasteurella multocida subsp.
multocida, Neisseria canis (based on 16S rRNA gene sequencing) and an Eikenella-like organism.
CHAPTER 42  Actinomycosis 401

soft tissue masses or draining skin lesions on the thoracic wall. accompanied by a thin body condition and fever. Dogs and cats
Sometimes there is a history of neck pain, gagging, or hypersali- with thoracic actinomycosis may show tachypnea or respiratory
vation before the onset of tachypnea, possibly due to passage of distress, and thoracic auscultation may reveal decreased lung
a penetrating foreign body. sounds if pyothorax or intrathoracic mass lesions are present.
Abdominal actinomycosis develops when ingested foreign Masses and/or ascites may be detected on abdominal palpation
bodies penetrate the gastrointestinal tract, which leads to the of dogs with abdominal actinomycosis. Dogs with retroperito-
formation of intra-abdominal mass lesions and ascites.2,16,23 neal actinomycosis can show pain on palpation of the abdomen
Abdominal involvement may also follow direct extension from or spine; signs of pelvic limb paralysis or paresis may be also
subcutaneous tissues or hematogenous spread of the organism present, such as pelvic limb ataxia, delayed or absent placing
to abdominal organs such as the liver. Although dogs are more reactions, increased segmental reflexes, and loss of pain sensa-
often affected than cats, intra-abdominal actinomycotic masses tion. Neurologic signs in dogs with Actinomyces meningitis or
can occur in cats.24,25 Retroperitoneal actinomycosis in dogs encephalitis include altered behavior, decreased consciousness,
often follows the migration of plant awns through the lung and cervical pain, vision loss, ataxia, tetraparesis, and seizures.
up the crus of the diaphragm to its dorsal attachment. Initially,
fever of unknown origin may be the only clinical sign. Ulti-
mately, progression can lead to osteomyelitis and compression
fractures of the vertebral bodies, with spinal pain and paresis or
paralysis of the pelvic limbs (Figure 42-3).3,26,27
Although rare, actinomycosis of the brain and meninges can
also occur in dogs (Figure 42-4).28-30 Brain abscesses may follow
hematogenous dissemination from a distant site, or meningitis
may result from extension of infection from an adjacent site,
such as the middle ear.29,31 In humans, risk factors for CNS
actinomycosis include dental disease or tooth extraction, head
trauma, gastrointestinal tract surgery, and chronic otitis, mas-
toiditis, or sinusitis. CNS actinomycosis has been reported in
dogs in association with concurrent otitis media/interna30 and
head trauma,28 and in cats in association with retrobulbar
abscesses31 and cutaneous abscesses of the tail base.13,32 FIGURE 42-4  Necropsy findings in a 1-year-old male neutered Yorkshire terrier with
Rarely, Actinomyces infections of the urinary bladder,12,33 suspected CNS actinomycosis. Three weeks previously, surgery to correct an extrahepatic
portosystemic shunt and remove a urate cystolith had been performed. The dog’s appetite
gallbladder,34,35 and heart valve4,10,36,37 have been recognized in
and activity level did not improve after the surgery, and for the week before the dog was
dogs, cats, and human patients. reexamined, disorientation and weakness was noted. The dog deteriorated rapidly after it
was hospitalized, and euthanasia was performed. Three separate abscesses were found in
Physical Examination Findings the left cerebral cortex. Histopathology revealed neutrophilic inflammation, hemorrhage,
Lesions in dogs and cats with cervicofacial or cutaneous-sub- and granular mats of material that contained gram-positive, acid-fast negative branching
cutaneous actinomycosis may fluctuant or firm, indurated, bacteria (consistent with Actinomyces spp.), as well as a dense population of gram-nega-
have draining sinuses, and rarely are ulcerated. Pain and fever tive, Giemsa-positive cocci. (Courtesy University of California, Davis, Veterinary Anatomic
are variable. Thoracic or abdominal actinomycosis is often Pathology Service.)

FIGURE 42-3  Left lateral spinal radiograph from a 3-year-old female Border collie with retroperitoneal actinomycosis. The dog was seen for a 3-day history of lethargy, fever, abdomi-
nal pain, and progressive pelvic limb paresis. Severe, smoothly marginated ventral spondylosis bridges the L1-L4 space. There is also marked irregular osteolysis of the 2nd, 3rd, and 4th
lumbar vertebrae. There is loss of visualization, narrowing, and irregularity of the intervertebral disc spaces at L2-3 and L3-4. There is marked loss of detail within the retroperitoneal space,
and the colon is displaced slightly ventrally. Irregular mineral foreign bodies are present in the stomach and colon.
402 SECTION 2  Bacterial Diseases

Diagnosis Cytologic Examination of Body Fluids


Cytologic examination of aspirates of abscesses, effusions, or
In the human literature, actinomycosis has been referred to CSF from animals with actinomycosis typically reveals a suppu-
as the “most misdiagnosed disease.”38 Fibrous masses are fre- rative to pyogranulomatous inflammatory response (>75% neu-
quently mistaken for neoplastic lesions, although reports of trophils). Total protein in pleural fluid is generally greater than
Actinomyces spp. infections that complicate neoplasia exist in 3.0 g/dL, with erythrocyte and nucleated cell counts often greater
human medicine.39,40 Aspiration of firm lesions is often unre- than 70,000 cells/µL. CSF from dogs and cats with cerebral acti-
warding, organisms are often difficult to isolate from lesions, nomycosis can grossly resemble pus. Aspirates of firm masses
and short periods of antimicrobial drug treatment are ineffec- may yield only a small amount of blood. Sulfur granules may
tive, which all serve to further mislead the clinician to a diag- be visible in effusion fluid or purulent material from cutaneous
nosis of neoplasia. Actinomycosis should be considered on lesions, grossly as white to tan to gray granules, or microscopi-
the list of differential diagnosis in any animal with a history cally as dense clusters of organisms. When bacteria are visual-
of penetrating plant awn foreign bodies, draining skin lesions, ized, they may be filamentous rods suggestive of Actinomyces
chronic fibrous masses, persistent bite wounds, pyothorax, spp., or “companion” microorganisms. Actinomycetes are gram-
retroperitoneal abscesses, or focal neurologic signs, especially positive, non-acid-fast filamentous organisms that are occasion-
when young-adult to middle-aged dogs are affected. Ultimately, ally branched. The filaments are less than 1 µm wide, vary in
diagnosis is based on clinical signs, identification of organisms length, and can stain irregularly, producing a beaded appearance
with typical morphology with cytology or histopathology, and (see Figure 42-1). Nocardia spp., Corynebacterium spp., and
culture. Mycobacterium spp. can be confused with Actinomyces species.

Laboratory Abnormalities Diagnostic Imaging


Complete Blood Count, Serum Biochemical Tests, and Urinalysis Plain Radiography
Animals with extensive, chronic actinomycosis may have mild Radiographs of cervicofacial or cutaneous-subcutaneous lesions
to moderate nonregenerative anemia, leukocytosis with a mild may show evidence of adjacent osteomyelitis and/or periosteal
to moderate left shift and monocytosis, hypoalbuminemia, and new bone formation. Pulmonary involvement may manifest as
hyperglobulinemia, which may be marked. Dogs with body cav- interstitial or alveolar infiltrates, sometimes with consolidation
ity effusions may be hypoglycemic. There are usually no specific (Figure 42-5, A). Air bronchograms may be seen within pulmo-
urinalysis findings. nary mass lesions, which suggests the presence of a nonneoplastic

A C
FIGURE 42-5  Imaging findings in a 1-year-old female spayed Labrador retriever with systemic actinomycosis secondary to grass awn migration. A, Dorsoventral thoracic radiograph.
Patchy diffuse interstitial opacities were present in all lung fields. B, Ultrasound image of the liver. Within the left liver, there were focal, ill-defined heteroechoic or hypoechoic nodules. One
of these areas (shown) had a hyperechoic rim with echogenic luminal contents. The nodules were consistent with multifocal hepatic abscesses. C, Ultrasound image of the thoracic wall.
A linear plant awn foreign body was identified (arrow).
CHAPTER 42  Actinomycosis 403

process. Other variable findings include pleural thickening, pleu- inflammation with or without the presence of filamentous bac-
ral effusion, widening of the mediastinum, pleural mass lesions, teria) and the presence of companion microorganisms support
enlargement of the cardiac silhouette (with pericardial involve- the diagnosis of actinomycosis.
ment), and periosteal new bone formation or osteomyelitis Specimens for culture can be collected through fine-needle
involving adjacent ribs, vertebral bodies, or sternebrae. Dogs aspiration or, if possible, biopsy. Because Actinomyces species
with retroperitoneal actinomycosis may have periosteal new are susceptible to many antimicrobial drugs, treatment of ani-
bone formation on the ventral aspects of two or three adjacent mals before obtaining specimens for culture can also prevent
vertebral bodies (usually T13 through L4) on abdominal radi- recovery of the organisms. Culture may be negative or yield only
ography; involvement of disc spaces is uncommon (see Figure “companion” microorganisms, which can obscure the presence
42-3). Abdominal actinomycosis may be characterized by loss of of Actinomyces spp. As a result, diagnosis is often based only
abdominal detail due to peritoneal effusion and intra-abdominal on cytologic or histologic identification of the organism in speci-
mass lesions. mens from animals with appropriate clinical signs.
Most Actinomyces spp. that cause disease in dogs and
Sonographic Findings cats are facultative anaerobes, but a few (A. bovis, A. israelii,
Ultrasonography in dogs with actinomycosis can be useful to A. meyeri) are obligate anaerobes.5,7,9,43 The facultative anaer-
identify linear grass awn foreign bodies (see Figure 42-5).41,42 obes can grow under aerobic conditions, and some, such as
Sonographic abnormalities in dogs with abdominal disease A. viscosus, grow best in these conditions. In addition, “com-
include variable amounts of flocculent ascites fluid, abdominal panion” microorganisms are often obligate anaerobes or other
lymphadenopathy, and nodular or mass lesions that incorporate aerobes. Therefore, both aerobic and anaerobic cultures should
or displace adjacent structures. New bone formation may be be requested when an Actinomyces spp. infection is suspected,
detected on interrogation of the ventral aspects of the vertebral and the laboratory should be notified that Actinomyces spp.
bodies in dogs with retroperitoneal disease. may be present. Tissue specimens, aspirates of pus, or sulfur
granules are ideal specimens for anaerobic culture.
Advanced Imaging Visible growth of Actinomyces spp. can occur within 48 hours
For animals with CNS involvement, MRI may reveal meningeal but usually requires 5 to 7 days. It may be necessary to hold
thickening, contrast enhancement, and/or intraparenchymal plates 2 to 4 weeks. Species identification using traditional bio-
contrast-enhancing mass lesions.18 MRI may also be useful for chemical tests is difficult. PCR amplification and DNA sequence
detection of grass awn foreign bodies. Advanced imaging allows analysis of the 16S rRNA gene may be necessary for precise spe-
assessment of the extent of actinomycosis (see Figure 42-2), cies identification. In the future, matrix-assisted laser desorption/
which can assist with surgical planning. ionization-time of flight (MALDI-TOF) mass spectrometry may
prove most useful for identification of Actinomyces species (see
Microbiologic Tests Chapter 3).44
Diagnostic assays available for Actinomyces infection in dogs In addition to actinomycetes, one to five other associated
and cats are shown in Table 42-1. bacteria are often recovered from properly handled specimens.
The most commonly isolated organisms are resident flora of the
Isolation and Identification oral cavity or intestinal tract and include the anaerobes Fuso-
Because Actinomyces is a commensal of the oral cavity, it is bacterium, Peptostreptococcus, Prevotella, or Bacteroides spp.;
commonly swallowed, inhaled, and transferred by licking; there- and Pasteurella multocida, Escherichia coli, and Streptococcus
fore, culture of the organism from the airways, gastrointestinal spp. Occasionally, pure cultures of Actinomyces spp. are iso-
tract, or skin does not necessarily constitute infection. Consis- lated, but this does not exclude the concurrent presence of other
tent cytologic or histopathologic findings (pyogranulomatous organisms, especially strict anaerobes.

TABLE 42-1
Diagnostic Assays Available for Actinomyces Infection in Dogs and Cats
Assay Specimen Type Target Performance
Culture Aspirates of purulent material Actinomyces spp. Some organisms require anaerobic conditions.
(e.g., abscesses, pleural effu- Requires several weeks’ incubation, and over-
sion), whole blood, CSF, tissue growth by companion bacteria may occur.
specimens obtained by biopsy Because Actinomyces spp. are mucosal com-
or at necropsy (e.g., subcutane- mensal bacteria, isolation from contaminated
ous tissue, heart valve, lung sites does not imply disease causation. PCR
tissue), sulfur granules and sequencing may be required to identify
the species present.
Cytology or histopa- Aspirates or tissue specimens Actinomyces spp. Organisms are gram positive, do not take up
thology with organ- organisms acid-fast stain, and are often accompanied by
ism detection using other bacteria and sulfur granules. Definitive
special stains diagnosis requires culture because Nocardia
can resemble Actinomyces spp.
404 SECTION 2  Bacterial Diseases

A B
FIGURE 42-6  Histopathology of a biopsy specimen from the xiphoid of a 10-year-old intact male English pointer dog with actinomycosis due to a migrating plant awn foreign body.
The dog had a 1-month history of a ventral thoracic wall mass. A, Dense, granular mats of branching bacteria were present in a background of extensive granulation tissue and embedded
plant material (not shown). H&E stain. B, Brown and Benn (gram) stain revealed colonies of gram-positive, filamentous, branching bacteria. Actinomyces bowdenii (based on 16S rRNA
gene sequencing), Pasteurella canis, Porphyromonas canoris, Propionibacterium acnes, a Bacteroides gingivalis–like organism, and a Bacteroides ureolyticus–like organism were isolated
from the lesion.

Molecular Diagnosis Using the Polymerase Chain Reaction Other nonfilamentous bacteria and/or plant material may also
Specific PCR assays for Actinomyces spp. have been developed be present. Actinomyces are not acid fast. With the rare excep-
and used for diagnosis of human actinomycosis45 but are not tion of some Nocardia spp. that are not acid fast, other fungi
commercially available for veterinary diagnosis. and bacteria that produce tissue granules can be distinguished
from Actinomyces based on their staining and morphologic
Pathologic Findings properties.
Gross lesions in animals with actinomycosis often consist of
one or more poorly defined, indurated masses of the subcu- Treatment and Prognosis
taneous tissues, thoracic cavity, lungs, abdominal cavity, or
retroperitoneal space, which incorporate adjacent structures. Treatment of actinomycosis usually requires prolonged antimi-
Masses may contain pockets of a reddish-brown exudate. crobial drug treatment. Penicillin is the antimicrobial drug of
Fistulas, plant material, and sulfur granules may be found. choice because isolates are uniformly susceptible to penicillins
Abdominal or thoracic effusions are often reddish-brown and (Table 42-2). High-dose therapy for weeks to months is gener-
can contain sulfur granules. Animals with thoracic and abdom- ally required for animals with chronic infections and extensive
inal infections may also have a diffuse, red, velvety to granular fibrosis. In human patients with actinomycosis, high doses of
thickening of the pleura or peritoneum and omentum. Rarely, penicillin are given parenterally for 2 to 6 weeks, followed by
abscesses are present in the brain, or purulent meningitis is oral therapy with amoxicillin for 6 to 12 months.48 If the animal
identified. is stable clinically, oral therapy can be tried from the outset.8,49
Histopathology of affected tissues reveals abscesses with a Treatment must be extended for weeks to months beyond reso-
core of neutrophils encapsulated by granulation tissue that con- lution of measurable disease to prevent relapse; in some cases,
tains macrophages, plasma cells, and lymphocytes in a dense, treatment for longer than a year may be required.8,16 However,
fibrous tissue matrix. When present, sulfur granules are gen- when actinomycosis is associated with foreign body migration,
erally in the center of microabscesses, but multiple tissue sec- shorter durations of treatment may be possible if the foreign
tions may be needed to find them. In tissue sections stained body is removed, drainage is established, and extensive scar
with hematoxylin and eosin, the granules are round, oval, or tissue has not formed. Alternative antimicrobial drug choices
scalloped amphophilic solid masses (Figure 42-6). They vary include clindamycin, doxycycline, chloramphenicol, carbap-
in size from 30 to 3000 µm in diameter and often are rimmed enems, and ceftriaxone.48 It is recommended that cephalexin,
by partially confluent radiating eosinophilic club-shaped struc- metronidazole, and aminoglycosides be avoided for treatment
tures collectively known as the Splendore-Hoeppli phenom- of human actinomycosis.48 Infections associated with com-
enon. This phenomenon can also occur with zygomycosis, panion microbes also usually resolve with penicillin, but on
sporotrichosis, parasitic infections, foreign body reactions, and occasion they require broader spectrum antibiotics during the
hypereosinophilic syndrome.46,47 Antigen-antibody complexes, initial treatment period followed by long-term administration
complement, and major basic protein of eosinophils have been of penicillin.
detected within the Splendore-Hoeppli phenomenon. Special Excepting when foreign body material is present (Figure 42-7),
stains (Gram stains, Giemsa and silver stains) are required to surgery has a controversial role in the treatment of actinomyco-
stain Actinomyces spp. and reveal clumps of tangled, intermit- sis, but may be required if there is extensive fibrous tissue for-
tently branched, thin (<1 µm) filaments (see Figure 42-6, B). mation or large accumulations of purulent material to facilitate
CHAPTER 42  Actinomycosis 405

TABLE 42-2
Medications That Could Be Used to Treat Actinomyces spp. Infections in Dogs and Cats with Actinomycosis
Drug Dose Route Interval (hours)
Penicillin G 100,000 U/kg IV, IM 6-8
Ampicillin sodium 20 IV, IM, SC 6-8
Amoxicillin 20 PO 8
Clindamycin* 11 PO 12 (dogs), 24 (cats)
Doxycycline† 5-10 PO 12-24

*For IV administration of clindamycin phosphate, see Chapter 8.


†10 mg/kg q12h preferred if tolerated without gastrointestinal adverse effects. Doxycycline hyclate or clindamycin should always be administered

with a bolus of water to avoid esophagitis.

period of antibiotic therapy may reduce the size of lesions and


improve lesion definition, which facilitates surgery followed by
continued antimicrobial drug treatment at a later date. Surgery
is also required if a nidus such as a grass awn is present.
Appropriate treatment of actinomycosis results in a cure rate
of greater than 90%.8,16,50 Cure rates with meningitis/meningo-
encephalitis and advanced thoracic or peritoneal disease with
extensive mass lesions may be lower.

Prevention
Prevention of actinomycosis involves restriction of access to
areas with extensive grass awn formation and avoidance of
fighting and biting activity between animals. When wounds
occur, they should be carefully examined for foreign material
and cleaned promptly. Plant awns found on animals’ coats
FIGURE 42-7  One-year-old female spayed Labrador retriever with systemic actino- should be removed and discarded as soon as possible and the
mycosis secondary to a plant awn foreign body after surgical removal of the plant awn and finding of one should prompt a thorough search for others.
placement of a grenade drain.
Public Health Aspects
drug penetration (see Chapter 87 for information on manage-
ment of pyothorax). In human medicine, an initial attempt to No reports exist of actinomycosis being transmitted from clini-
control disease with aggressive medical therapy alone has been cally infected animals to humans or other animals; however,
suggested, with surgical therapy if the response to treatment is humans bitten by dogs, cats, or other people can develop acti-
inadequate.48 In animals with pulmonary abscesses, removal of nomycosis (see Chapter 57).51 The most common species that
affected lung lobes may be required. Fibrotic lesions are often infects humans is A. israelii.48 This is a commensal of the canine
extensively vascularized and may obliterate tissue planes, which oral cavity but has not been isolated from dogs or cats with
complicates dissection. In dogs with solitary masses that involve actinomycosis. Nevertheless, A. viscosus, A. odontolyticus, and
the thoracic and abdominal walls, radical surgical excision is A. meyeri have been isolated from canine and feline as well as
often curative, but repeat surgeries may be needed. An initial human actinomycosis.

CASE EXAMPLE in thirst or urination. Earlier the same day, Penny had been
evaluated at another veterinary clinic where a grade IV/
Signalment: “Penny”, a 1-year-old, female spayed Labrador VI left-sided cardiac murmur was auscultated and thoracic
retriever from Herald in northern California radiographs showed a bronchoalveolar and interstitial lung
History: Penny was evaluated by the University of California, pattern. She was treated with cefazolin (27 mg/kg IV) and
Davis, veterinary emergency service for a 1-week history of furosemide (1 mg/kg SC) and referred. Penny had been fully
progressive inappetence, weight loss, lethargy, and fever. vaccinated for canine distemper virus, canine parvovirus,
Two days before she was evaluated, Penny vomited up some canine adenovirus, and rabies. She had missed some doses
kibble, and had not eaten anything since. The owner reported of heartworm prophylaxis medication recently and received
that her feces had been formed and there were no changes no other medications. She lived with three other healthy
406 SECTION 2  Bacterial Diseases

Labrador retrievers. She roamed on 8 acres of land, and Urinalysis: SGr 1.016; pH 5.0, 1+ protein (SSA), 1+ bilirubin,
there was a pond on the property where she swam and was 2+ hemoprotein, 1+ glucose, 0-1 WBC/HPF, 0-3 RBC/HPF,
known to eat crayfish and frogs. Her usual diet consisted of rare granular casts, many amorphous crystals, rare bilirubin
a commercial dry puppy food. Seven months earlier, she had crystals, many lipid droplets
been evaluated at another veterinary clinic for vomiting and Coagulation Panel: PT 7.7 s (7.5-10.5 s), APTT 15.2 s (9-12 s),
inappetence. At that time she was moderately azotemic; fibrinogen 232 mg/dL (90-255 mg/dL), D-dimer 1.0-2.0 µg/
an exploratory laparotomy was performed for a suspected mL (0-0.25 µg/mL).
foreign body, but no foreign body was found. Imaging Findings:
Physical Examination: Thoracic Radiographs (3 View): There were patchy diffuse
Body Weight: 22.5 kg unstructured interstitial opacities in all lung lobes (see
General: Quiet, alert, responsive and wagging tail. Figure 42-5, A). Cardiovascular structures appeared within
Approximately 5% dehydrated. T = 105.2°F (40.7°C), HR 160 normal limits. The liver was markedly enlarged.
beats/min, RR = 42 breaths/min. Pink, slightly tacky mucous Echocardiography: The left ventricle was eccentrically
membranes, CRT < 2 s. hypertrophied. The papillary muscles had normal shape and
Integument: Full clean haircoat, no evidence of ectoparasites. echogenicity. There was no enlargement of the left atrium.
Eyes, Ears, Nose, and Throat: No clinically significant There was no evidence of endocarditis. Decreased fractional
abnormalities were noted. shortening (26%; reference range, approximately 34%-46%)
Musculoskeletal: BCS 3/9. Generalized mild muscle atrophy and increased end systolic diameter (3.9 cm; reference range,
was present. approximately 1.8-3.5 cm) and E-point septal separation (6.8
Cardiovascular: A grade II/VI left systolic murmur was mm) were indicative of systolic dysfunction, which could
auscultated. Femoral pulses were strong, regular, and also be appreciated subjectively. There was mild tricuspid
synchronous. regurgitation and trivial pulmonic insufficiency. Conclusions:
Respiratory: Increased breath sounds were present bilaterally Moderate systolic dysfunction. It was recommended that
and diffusely. There was a mild increase in respiratory effort. the dog’s diet and taurine blood/plasma levels be evaluated.
All Other Systems: No clinically significant abnormalities were It was also considered possible that the dog’s febrile state
noted. was responsible for some of abnormalities observed on the
Laboratory Findings: echocardiogram.
CBC: Abdominal Ultrasound: The liver was diffusely mottled,
HCT 24.0% (40%-55%), particularly in the left caudal region (see Figure 42-5, B).
MCV 65.6 fL (65-75 fL) Within the left caudal liver, there was a focal, ill-defined
MCHC 35.0 g/dL (33-36 g/dL) heteroechoic area approximately 1 cm in diameter. Other
WBC 29,850 cells/µL (6000-13,000 cells/µL) smaller, ill-defined hypoechoic nodules were also present
Neutrophils 16,119 cells/µL (3000-10,500 cells/µL) within the left liver. One of these areas had a well-demarcated,
Lymphocytes 2985 cells/µL (1000-4000 cells/µL) hyperechoic rim with irregular echogenic luminal contents
Monocytes 7761 cells/µL (150-1200 cells/µL) and measured approximately 0.5 cm in diameter. Multiple
Eosinophils 0 cells/µL (0-1500 cells/µL) large mesenteric lymph nodes were just caudal to the liver;
Basophils 0 cells/µL (0-50 cells/µL) the largest measured 1.3 cm by 4.5 cm. There was a small
Platelets 98,000 platelets/µL (150,000-400,000 platelets/µL) amount of anechoic free abdominal fluid. The spleen was
MPV 20.1 fL (7-13 fL). moderately enlarged, but the echotexture and echogenicity
A few slightly toxic band neutrophils and moderate appeared within normal limits. There was mild renal
numbers of macroplatelets were present. pyelectasia bilaterally, but the renal papillae appeared within
Serum Chemistry Profile: normal limits.
Sodium 144 mmol/L (145-154 mmol/L) Microbiologic Testing: Aerobic and anaerobic blood
Potassium 4.3 mmol/L (3.6-5.3 mmol/L) cultures (five specimens): Negative
Chloride 114 mmol/L (108-118 mmol/L) IFA serology for serum antibody to Ehrlichia canis, Anaplas-
Bicarbonate 15 mmol/L (16-26 mmol/L) ma phagocytophilum, Rickettsia rickettsii, and Babesia ca-
Phosphorus 5.3 mg/dL (3.0-6.2 mg/dL) nis: Negative
Calcium 9.6 mg/dL (9.7-11.5 mg/dL) IFA serology for serum antibody to Bartonella vinsonii subsp.
BUN 22 mg/dL (5-21 mg/dL) berkhoffii, Bartonella clarridgeiae, and Bartonella hense-
creatinine 0.8 mg/dL (0.3-1.2 mg/dL) lae: Negative
Glucose 44 mg/dL (64-123 mg/dL) Bartonella culture (whole blood): Negative
Total protein 7.8 g/dL (5.4-7.6 g/dL) Serology for Coccidioides serum antibodies: Negative
Albumin 2.5 g/dL (3.0-4.4 g/dL) Cytologic Findings:
Globulin 5.3 g/dL (1.8-3.9 g/dL) Splenic Aspirate (Ultrasound-Guided): Two smears were
ALT 31 U/L (19-67 U/L) examined that contained a large amount of blood in
AST 76 U/L (19-42 U/L) the background and were highly cellular. Several small
ALP 612 U/L (21-170 U/L) clumps of splenic stromal cells were noted. A large
Gamma GT 5 U/L (0-6 U/L) heterogeneous population of lymphocytes was found,
Cholesterol 292 mg/dL (135-361 mg/dL) with a moderate increase in plasma cells. A moderate
Total bilirubin 0.8 mg/dL (0-0.2 mg/dL) number of hematopoietic cells was also present, including
CHAPTER 42  Actinomycosis 407

megakaryocytes, myeloid, and erythroid precursors. showed persistent hypoechogenicity of the left liver lobe,
Interpretation: Moderate reactive lymphoid hyperplasia and but during the procedure a hyperechoic lesion that was
extramedullary hematopoiesis. consistent with a plant awn foreign body was discovered
Liver Aspirate (Ultrasound-Guided): Smears contained a in the subcutaneous tissue in a caudoventral intercostal
diffusely stippled background with abundant nuclear debris space. On day 6, surgery was performed to remove the
and were highly cellular. A large population of mildly to plant awn. Packed red cells were administered during
markedly degenerate neutrophils were observed. Degenerate the procedure, and a 16F thoracostomy tube was placed
cells ranged from highly pyknotic to severely karyolytic. because the thoracic cavity was entered during the search
A few neutrophils contained small intracellular bacterial for the foxtail, which, with the assistance of intraoperative
rods. Interpretation: Marked suppurative inflammation with ultrasound examination, was eventually located under the
bacterial sepsis. 13th rib just outside the pleural space. Sulfur granules were
Further Microbiologic Testing: found embedded in the tissue during surgery. A closed
Aerobic and Anaerobic Culture (Ultrasound-Guided Liver suction grenade drain was also placed (see Figure 42-7). The
Aspirate): Small numbers of suspect Actinomyces spp. thoracostomy tube was removed later in the day. Treatment
No anaerobes cultured. Partial 16S rRNA gene PCR and with enrofloxacin and ampicillin was continued, as well as
sequence analysis revealed 99% identity to Actinomyces with oxymorphone (0.04 mg/kg SC q6h) to control pain.
bowdenii. On day 7, Penny’s temperature was 100.7°F (38.2°C), and
Diagnosis: Systemic A. bowdenii infection with hepatic the drain was removed. Aerobic and anaerobic culture of
involvement the foxtail revealed small numbers of penicillin-susceptible
Treatment: Immediately on admission, Penny was treated Enterococcus faecalis and an organism resembling a
with IV lactated Ringer’s solution with 20 mEq/L KCl and 5% Streptomyces sp. No anaerobes were cultured. Penny was
dextrose at 75 mL/hr. After the specimens for blood culture discharged on day 8 with instructions to continue treatment
were collected, treatment with enrofloxacin (10 mg/kg with clavulanic acid–amoxicillin for an additional month. The
IV q24h) and ampicillin (22 mg/kg IV q8h) was initiated. dog had no further clinical signs of illness.
Within 24 hours, the dog’s rectal temperature had decreased Comments: This is an unusual case of disseminated A. bowdenii
to 101.3°F (38.5°C), the blood glucose had increased from infection that led to hematogenous pneumonia and hepatic
41 mg/dL to 107 mg/dL, and Penny began eating. The liver abscessation. Although there was an initial defervescence
aspirate was submitted for culture on day 2 of hospitalization. with antimicrobial drug treatment, anemia and pyrexia
A recheck abdominal ultrasound examination on day 3 persisted, which led to a search for an abscess that might
showed progressive abscessation of the left liver and further require drainage or a plant awn. Fortunately, a plant awn
enlargement of the gastrohepatic lymph nodes. By day was ultimately identified with ultrasound and removed, after
4 of hospitalization, growth of an organism resembling which the dog’s illness resolved completely. E. faecalis was
an Actinomyces sp. was reported. On day 5, antimicrobial isolated from the plant awn and represented a “companion
drug treatment was changed to clavulanic acid–amoxicillin microbe,” but whether dissemination of this organism
(17 mg/kg q8h). However, later in the day, Penny’s temperature occurred was unclear. The Streptomyces sp. may have been a
increased to 103.6°F (39.8°C), and so the initial antimicrobial contaminant, or the Actinomyces sp. that was misidentified.
drug prescription was reinstated. The dog’s hematocrit had The owner was warned that additional plant awns might
also dropped to 19%. Thoracic radiographs were repeated and still be present. The cause of the cardiomyopathy was not
showed almost complete resolution of the patchy pulmonary determined; plasma and whole blood taurine concentrations
infiltrates. A follow-up echocardiogram was unchanged. were within normal limits, and follow-up examination was
Abdominal ultrasound examination was again repeated and not performed.

SUGGESTED READINGS 4. Meurs KM, Heaney AM, Atkins CE, et  al. Comparison of poly-
merase chain reaction with bacterial 16S primers to blood culture
Barnes LD, Grahn BH. Actinomyces endophthalmitis and pneumonia in to identify bacteremia in dogs with suspected bacterial endocardi-
a dog. Can Vet J. 2007;48:1155-1158. tis. J Vet Intern Med. 2011;25:959-962.
Edwards DF, Nyland TG, Weigel JP. Thoracic, abdominal, and verte- 5. Pascual C, Foster G, Falsen E, et al. Actinomyces bowdenii sp. nov.,
bral actinomycosis. Diagnosis and long-term therapy in three dogs. isolated from canine and feline clinical specimens. Int J Syst Bacte-
J Vet Intern Med. 1988;2:184-191. riol. 1999;49(Pt 4):1873-1877.
Kirpensteijn J, Fingland RB. Cutaneous actinomycosis and nocardiosis in 6. Buchanan AM, Scott JL. Actinomyces hordeovulneris, a canine
dogs: 48 cases (1980-1990). J Am Vet Med Assoc. 1992;201:917-920. pathogen that produces L-phase variants spontaneously with coin-
cident calcium deposition. Am J Vet Res. 1984;45:2552-2560.
REFERENCES 7. Hoyles L, Falsen E, Foster G, et  al. Actinomyces coleocanis
1. Bollinger O. Über eine neue Pilmkrankheit beim Rinde. Zbl Med sp. nov., from the vagina of a dog. Int J Syst Evol Microbiol.
Wissensch. 1877;15:481-485. 2002;52:1201-1203.
2. Georg LK, Brown JM, Baker HJ, et al. Actinomyces ­viscosus as an 8. Edwards DF, Nyland TG, Weigel JP. Thoracic, abdominal, and
agent of actinomycosis in the dog. Am J Vet Res. 1972;33:1457-1470. vertebral actinomycosis. Diagnosis and long-term therapy in three
3. Davenport AA, Carter GR, Schirmer RG. Canine actinomycosis dogs. J Vet Intern Med. 1988;2:184-191.
due to Actinomyces viscosus: report of six cases. Vet Med Small 9. Hoyles L, Falsen E, Pascual C, et al. Actinomyces catuli sp. nov.,
Anim Clin. 1974;69:1442, 1444-1447. from dogs. Int J Syst Evol Microbiol. 2001;51:679-682.
408 SECTION 2  Bacterial Diseases

10. Junius G, Bavegems V, Stalpaert M, et  al. Mitral valve endo- 31. Barrs VR, Nicoll RG, Churcher RK, et al. Intracranial empyema:
carditis in a Labrador retriever caused by an actinomyces spe- literature review and two novel cases in cats. J Small Anim Pract.
cies identified as Actinomyces turicensis. J Vet Intern Med. 2007;48:449-454.
2004;18:899-901. 32. Stowater JL. Actinomycosis in the spinal canal of cat. Feline Pract.
11. Sykes JE, Unpublished observations, 2012. 1978;8:26-27.
12. Billington SJ, Post KW, Jost BH. Isolation of Arcanobacterium 33. Dhamborvorn T, Tritipsatit S, Meemongkoldilok S. Actinomycosis
(Actinomyces) pyogenes from cases of feline otitis externa and of the urinary bladder. J Med Assoc Thai. 2001;84:109-112.
canine cystitis. J Vet Diagn Invest. 2002;14:159-162. 34. Ormsby AH, Bauer TW, Hall GS. Actinomycosis of the cholecystic
13. Bestetti G, Bühlman V, Nicolet J, et  al. Paraplegia due to Acti- duct: case report and review. Pathology. 1998;30:65-67.
nomyces viscosus infection in a cat. Acta Neuropathol (Berl). 35. Harvey AM, Holt PE, Barr FJ, et  al. Treatment and long-term
1977;39:231-235. follow-up of extrahepatic biliary obstruction with bilirubin choleli-
14. Love DN, Jones RF, Bailey M, et al. Isolation and characterisation thiasis in a Somali cat with pyruvate kinase deficiency. J Feline Med
of bacteria from abscesses in the subcutis of cats. J Med Microbiol. Surg. 2007;9:424-431.
1979;12:207-212. 36. Sykes JE, Kittleson MD, Pesavento PA, et al. Evaluation of the rela-
15. Love DN, Jones RF, Bailey M, et al. Isolation and characterisation tionship between causative organisms and clinical characteristics of
of bacteria from pyothorax (empyaemia) in cats. Vet Microbiol. infective endocarditis in dogs: 71 cases (1992-2005). J Am Vet Med
1982;7:455-461. Assoc. 2006;228:1723-1734.
16. Hardie EM, Barsanti JA. Treatment of canine actinomycosis. J Am 37. Mardis JS, Many Jr WJ. Endocarditis due to Actinomyces viscosus.
Vet Med Assoc. 1982;180:537-541. South Med J. 2001;94:240-243.
17. Kirpensteijn J, Fingland RB. Cutaneous actinomycosis and nocar- 38. Cope VZ. Visceral actinomycosis. Ann R Coll Surg Engl.
diosis in dogs: 48 cases (1980-1990). J Am Vet Med Assoc. 1949;5:394-410.
1992;201:917-920. 39. Afolabi IR, Shashidhar VM. Carcinoma of the oesophagus mas-
18. Sykes JE. Actinomycosis and nocardiosis. In: Greene CE, ed. Infec- querading as actinomycosis: a case report and a review of litera-
tious Diseases of the Dog and Cat. 4th ed. St. Louis: Elsevier Saun- ture. Pac Health Dialog. 2004;11:94-95.
ders; 2012:484-495. 40. Batur Calis A, Ozbal AE, Basak T, et al. Laryngeal actinomycosis
19. Love DN, Jones RF, Bailey M, et al. Bacteria isolated from subcu- accompanying laryngeal carcinoma: Report of two cases. Eur Arch
taneous abscesses in cats. Aust Vet Pract. 1978;8:87-90. Otorhinolaryngol. 2006;263:783-785.
20. Ochiai K, Kurita-Ochiai T, Kamino Y, et  al. Effect of co- 41. Frendin J. Diagnostic imaging of back pain. J Small Anim Pract.
aggregation on the pathogenicity of oral bacteria. J Med Microbiol. 1999;40:278-285.
1993;39:183-190. 42. Sivacolundhu RK, O’Hara AJ, Read RA. Thoracic actinomycosis
21. Ledbetter EC, Scarlett JM. Isolation of obligate anaerobic bacte- (arcanobacteriosis) or nocardiosis causing thoracic pyogranuloma
ria from ulcerative keratitis in domestic animals. Vet Ophthalmol. formation in three dogs. Aust Vet J. 2001;79:398-402.
2008;11:114-122. 43. Hoyles L, Falsen E, Pascual C, et al. Actinomyces catuli sp. nov.,
22. Barnes LD, Grahn BH. Actinomyces endophthalmitis and pneumo- from dogs. Int J Syst Evol Microbiol. 2001;51:679-682.
nia in a dog. Can Vet J. 2007;48:1155-1158. 44. Ng LS, Sim JH, Eng LC, et al. Comparison of phenotypic methods
23. Chastain CB, Grier RL, Hogle RM, et al. Actinomycotic peritonitis and matrix-assisted laser desorption ionisation time-of-flight mass
in a dog. J Am Vet Med Assoc. 1976;168:499-501. spectrometry for the identification of aero-tolerant Actinomyces
24. Kawamura N, Shimada A, Morita T, et al. Intraperitoneal actino- spp. isolated from soft-tissue infections. Eur J Clin Microbiol Infect
mycosis in a cat. Vet Rec. 2005;157:593-594. Dis. 2011.
25. Sharman MJ, Goh CS, Kuipers von Lande RG, et al. Intra-abdominal 45. Fujita Y, Iikura M, Horio Y, et al. Pulmonary Actinomyces graeve-
actinomycetoma in a cat. J Feline Med Surg. 2009;11:701-705. nitzii infection presenting as organizing pneumonia diagnosed by
26. Frendin J, Greko C, Hellmén E, et al. Thoracic and abdominal wall PCR analysis. J Med Microbiol. 2012;61:1156-1158.
swellings in dogs caused by foreign bodies. J Small Anim Pract. 46. Read RW, Zhang J, Albini T, et  al. Splendore-Hoeppli phenom-
1994;35:499-508. enon in the conjunctiva: immunohistochemical analysis. Am J Ophthal-
27. Johnson DE, Summers BA. Osteomyelitis of the lumbar verte- mol. 2005;140:262-266.
bral in dogs caused by grass-seed foreign bodies. Aust Vet J. 47. Sykes JE, Weiss DJ, Buoen LC, et al. Idiopathic hypereosinophilic
1971;47:289-294. syndrome in 3 Rottweilers. J Vet Intern Med. 2001;15:162-166.
28. Anvik JO, Lewis R. Actinomyces encephalitis associated with 48. Russo TA. Agents of actinomycosis. In: Mandell GL, Bennett JE,
hydrocephalus in a dog. Can Vet J. 1976;17:42-44. Dolin R, eds. Principles and Practice of Infectious Diseases. 7th ed.
29. Couto SS, Dickinson PJ, Jang S, et  al. Pyogranulomatous menin- Philadelphia, PA: Elsevier; 2010:3209-3219.
goencephalitis due to Actinomyces sp. in a dog. Vet Pathol. 49. Nelson JD, Hermann DW. Oral penicillin therapy for thoracic acti-
2000;37:650-652. nomycosis. Pediatr Infect Dis. 1986;5:594-595.
30. Sturges BK, Dickinson PJ, Kortz GD, et al. Clinical signs, magnetic 50. Frendin J. Pyogranulomatous pleuritis with empyema in hunting
resonance imaging features, and outcome after surgical and medi- dogs. Zentralbl Vet Med. 1997;44:167-178.
cal treatment of otogenic intracranial infection in 11 cats and 4 51. Reiner SL, Harrelson JM, Miller SE, et al. Primary actinomycosis of an
dogs. J Vet Intern Med. 2006;20:648-656. extremity: a case report and review. Rev Infect Dis. 1987;9:581-589.
CHAPTER 43

Nocardiosis
Jane E. Sykes

spp. are ubiquitous soil saprophytes that degrade organic mat-


Overview of Nocardiosis ter. They are also found in fresh and salt water, in dust, and on
First Described: 1888, Alfort, France (Edmond Nocard)1 decaying plants and fecal matter.2 They can be carried mechani-
cally on the claws or skin. Infections are acquired via inhalation
Causes: Nocardia spp. (order Actinomycetales, family
of organisms or inoculation via puncture wounds. Transfer of
Nocardiaceae)
infection between animals does not occur.
Geographic Distribution: Worldwide, although infections in Nocardiosis is much less frequently reported in dogs and cats
dogs and cats are most commonly reported from Brazil, than actinomycosis, although nocardiosis is increasingly recog-
the western United States, and Australia nized in both human medicine and companion animals in asso-
Mode of Transmission: Inhalation or cutaneous inoculation of ciation with immunocompromise, such as occurs with potent
organisms in the environment, with opportunistic prolif- immunosuppressive drug therapy (especially cyclosporine).3-5
eration in the face of impaired host immune defenses Where vaccination for canine distemper virus (CDV) is inad-
equate or not widely instituted, nocardiosis in dogs has been
Major Clinical Signs: Lethargy, weight loss, inappetence;
associated with CDV-induced immunosuppression.6
subcutaneous masses and nonhealing, crusted, or drain-
The prevalence of nocardiosis and the predominant Nocar-
ing skin lesions; pulmonary nodules, masses and/or effu-
dia species that cause disease vary geographically. For example,
sion with cough and tachypnea; signs of dissemination
within the United States, human nocardiosis has been most
(neurologic signs, chorioretinitis, abdominal effusion or
commonly reported from the southwestern states. Dry, dusty,
masses, lameness)
and windy conditions in these areas may facilitate aerosoliza-
Differential Diagnoses: Actinomycosis, streptomycosis, tion and dispersal of nocardiae.7 Dogs and cats from Australia
mycobacteriosis, bartonellosis, fungal infections, neopla- and the western United States are most commonly infected with
sia, leishmaniosis Nocardia nova.8-10 Other species isolated from dogs and cats
Human Health Significance: Direct transmission of disease are shown in Table 43-1.11-16 In humans, Nocardia brasiliensis
from animals to humans does not occur, but humans have is the most common species isolated from patients that reside in
developed disease after bites or scratches from healthy tropical locations such as the southwestern United States, Cen-
dogs or cats. tral and South America, and Australia.
More than 60% of people with nocardiosis have underlying
immunosuppressive disorders such as AIDS, chronic obstructive
pulmonary disease, autoimmune diseases that require immuno-
Etiology and Epidemiology suppressive drug therapy, solid organ transplantation, diabetes

Nocardia are filamentous, branching, gram-positive bacteria


that belong to the family Nocardiaceae (Figure 43-1). More
than 50 Nocardia species have been recognized since the advent
of molecular identification methods, approximately half of
which are human and animal pathogens.2 The advent of molec-
ular methods has also led to extensive revision of the taxonomy
of Nocardia spp. Species that belong to the former N. aster-
oides complex are now considered distinct species and include
Nocardia cyriacigeorgica (formerly N. asteroides sensu stricto),
Nocardia abscessus, Nocardia nova, Nocardia farcinica, and
Nocardia otitidiscaviarum.2 Knowledge of the species involved
is clinically important, because each Nocardia species differs
in its antimicrobial susceptibility patterns, epidemiology, and
pathogenicity. They also differ in their cell wall mycolic acid
content, which affects their ability to stain with acid-fast stains.
Like actinomycosis, nocardiosis is a suppurative to granu-
lomatous, localized, or disseminated opportunistic infection
caused by filamentous bacteria. However, unlike Actinomyces FIGURE 43-1  Gram-stained smear showing Nocardia spp. The organisms are
spp. (which are commensals of mucous membranes), Nocardia ­gram-positive, filamentous, branching, and slightly beaded.

409
410 SECTION 2  Bacterial Diseases

mellitus, and hemic neoplasia.17 Underlying immunosuppres-


TABLE 43-1 sive disease or drug treatment also seems to predispose dogs to
Nocardia Species Isolated from Dogs and Cats with Nocardiosis nocardiosis. Nocardiosis has been documented in several dogs
treated with cyclosporine and in a dog with lymphoma.3-5,9
Host Species Dogs Country Most affected cats have cutaneous infections that develop after
scratch or bite wounds.8,18 Accordingly, more than 75% to
Dogs N. nova United States, 80% of affected cats are male; in the author’s hospital, 9 of 10
Australia8,9 affected cats were male.8,9,19 Cats of any age can be affected,
N. paucivorans United States9 and no breed predilection has been identified.8 Some cats have
N. asiatica United States9 underlying disorders that predispose them to nocardiosis, such
N. abscessus United States5 as a history of renal transplantation, underlying retroviral infec-
N. otitidiscaviarum Brazil6 tion, or glucocorticoid administration. Others have no obvious
N. farcinica Australia8 underlying immunosuppressive disease.
Cats N. nova United States,
Australia9 Clinical Features
N. cyriacigeorgica Australia8
N. africana Japan,11 Brazil12 Signs and Their Pathogenesis
N. elegans Japan13 The pathogenicity of Nocardia spp. is influenced by the strain
N. brasiliensis United States14 and growth phase of the organism, and host susceptibility.
N. otitidiscaviarum Spain15 Virulent Nocardia strains are facultative intracellular patho-
N. tenerifensis United States16 gens that inhibit phagosome-lysosome fusion, neutralize phago-
somal acidification, resist oxidative burst, secrete superoxide
dismutase, and alter lysosomal enzymes within neutrophils and
macrophages.17 These effects are partly related to the content
and structure of mycolic acids within the bacterial cell wall,
which vary among strains and throughout the growth phase.
Filamentous log-phase cells in the environment can be highly
resistant to phagocytosis. Some strains have a greater propen-
sity to invade the central nervous system (CNS).19,20
The normal host response to infection is characterized by
an initial pyogenic inflammatory response, but a cell-mediated
immune response is necessary to destroy the organisms. Dimin-
ished host resistance, especially impaired cell-mediated immu-
nity (CMI), is a primary factor in susceptibility to nocardiosis
and the extent to which dissemination occurs.
Cutaneous-subcutaneous nocardiosis, the most common
form in cats (>75% of affected cats), is characterized by slow
and progressive circumferential spread of a nonhealing, drain-
A ing wound (Figure 43-2). Lesions are typically subacute to
chronic. Infections of feeding tube sites can also occur.21 Myce-
toma-like lesions in the inguinal area resemble those described
for rapidly growing mycobacterial infections (see Chapter 44),
with multiple draining sinuses. These may result from contami-
nation of “raking” injuries inflicted by the hindlimbs during
fighting behavior,8 or contamination of traumatic wounds from
penetrating plant material. Cutaneous-subcutaneous nocardio-
sis was documented in 8 of 9 dogs from Brazil6 but represents
the minority of canine nocardiosis cases in California and Aus-
tralia.8,9 Dogs with cutaneous involvement typically develop
draining wounds and masses, often on the head and limbs.
Osteomyelitis can occur in association with cutaneous lesions.
The pathogenesis of pulmonary or disseminated nocardiosis
is similar to that of the deep mycoses. Branching nocardial fila-
ments fragment into small, unicellular particles that are aero-
solized and inhaled, possibly in dust. Once within the lung,
B Nocardia spp. proliferate in the face of immune suppression,
which leads to formation of intrapulmonary masses or pneu-
FIGURE 43-2  Six-year-old male neutered domestic shorthair cat with cutaneous-
subcutaneous nocardiosis. The lesions progressed over at least a year after a cat bite monia, hilar lymphadenopathy, and/or extrapulmonary masses
abscess that was initially treated with amoxicillin-clavulanic acid. They consisted of nod- and pyothorax (Figure 43-3, A). Pulmonary nocardiosis is the
ular and crusted lesions on the head (A) and cervical region (B) that drained fluid and most common form of nocardiosis in humans and also occurs in
extended down the cervical region to the cranioventral thorax. The cat tested negative dogs and less commonly in cats. In dogs, it can have a peracute
for retroviruses. (Courtesy University of California, Davis, Veterinary Dermatology Service.) onset characterized by tachypnea, hemoptysis, hypothermia,
CHAPTER 43  Nocardiosis 411

A B
FIGURE 43-3  Gross necropsy findings in a 4-year-old male neutered domestic shorthair cat with disseminated nocardiosis caused by Nocardia nova. The cat was a renal transplant
recipient. A, Multifocal to coalescing large, white to gray, caseous pulmonary masses are present within the lungs. B, The brain contained multifocal, white to gray foci that histologically
consisted of large numbers of neutrophils and filamentous bacteria. (Courtesy University of California, Davis Veterinary Anatomic Pathology Service.)

collapse, and death; however, subacute to chronic clinical signs with a history of immunosuppression, although the last is not
are more common.22-25 Co-infection with CDV is occasionally always present. Confirmation of the diagnosis requires culture.
reported.6,26,27 Disseminated nocardiosis occurs when organ-
isms in the lung erode into blood vessels and spread systemi- Laboratory Abnormalities
cally, with abscess formation in a variety of organs. This results Complete Blood Count and Serum Biochemical Tests
in signs of lethargy, fever, inappetence, and signs that relate Hematologic abnormalities in nocardiosis are similar to those
to the location of the infectious process. Disseminated disease with actinomycosis (nonregenerative anemia, neutrophilic leu-
occurs in cats as well as in dogs. The most frequently involved kocytosis with a left shift, monocytosis, and sometimes marked
extrathoracic organs after dissemination are skin and subcuta- hyperglobulinemia). However, in immunosuppressed animals,
neous tissue, kidney, liver, spleen, lymph nodes, CNS, eye, bone, lymphopenia, monocytopenia, and eosinopenia may be pres-
and joints. In dogs and cats, CNS lesions can result in seizures ent. Hypercalcemia associated with granulomatous disease was
or neurologic signs that relate to the local effects of abscesses or reported in a cat with nocardiosis.29
granulomas in the brain, meninges, or spinal cord (see Figure
43-3, B). CNS involvement is common in human nocardiosis. Cytologic Examination of Body Fluids
Peritoneal nocardiosis has rarely been reported in cats, possibly Examination of pleural effusion, bronchoalveolar lavage fluid,
as a result of penetrating wounds to the abdomen.8,28 and aspirates of abscesses from animals with nocardiosis typi-
cally reveals suppurative to pyogranulomatous inflammation,
Physical Examination Findings with large numbers of degenerate neutrophils. Gram-positive,
Physical examination findings in animals with cutaneous- often partially or weakly acid-fast, beaded, filamentous organ-
subcutaneous disease consist of chronic, sometimes crusted, isms that branch at right angles may be observed individually
ulcerated, nonhealing draining wounds, and in cats may or in loose aggregates (see Figure 43-1). The filaments may also
involve the extremities, inguinal area, flank, head, bridge of fragment to form rods and coccoid forms. In contrast, Acti-
the nose, and neck (see Figure 43-2). Animals with pulmonary nomyces spp. is not acid fast, and Mycobacterium spp. do not
or systemic involvement may have a thin body condition, leth- branch. When the infecting Nocardia sp. is not acid fast, it may
argy, and fever. Tachypnea and cough may be present, and not be distinguishable from an Actinomyces sp. organism. With
lung sounds may be increased (with bronchopneumonia) or Romanowsky-type stains, organisms are eosinophilic or baso-
decreased (with pyothorax). Involvement of the liver, spleen, philic and have a beaded appearance. They are usually around
and lymph nodes may result in hepatomegaly, splenomegaly, 0.5 µm in diameter and up to 30 µm long, but thicker organ-
and/or peripheral or abdominal lymphadenomegaly. Oph- isms were described in a dog infected with N. abscessus (2 µm).5
thalmic examination may reveal chorioretinitis. Bone or joint Macroaggregates (i.e., sulfur granules) occur infrequently in
infection results in focal limb swelling and lameness. Animals effusions. In contrast to actinomycosis, mixed bacterial popula-
with CNS involvement may show mental obtundation, aniso- tions in deep tissue sites are rarely present.
coria, decreased menace and pupillary light responses, head
tilt, nystagmus, decreased gag reflexes, and/or delayed or Diagnostic Imaging
absent placing reactions. Plain Radiography
Radiographs of cutaneous-subcutaneous lesions can reveal
Diagnosis soft tissue swelling with or without associated bone lysis and
periosteal proliferation. The radiographic appearance of pul-
A diagnosis of nocardiosis is usually suspected based on the monary lesions varies and includes multiple, diffuse pulmonary
presence of persistent pyogenic to pyogranulomatous inflamma- nodules; intrapulmonary or extrapulmonary solitary masses;
tory lesions and the presence of filamentous bacteria, together focal or diffuse bronchointerstitial to alveolar infiltrates; lobar
412 SECTION 2  Bacterial Diseases

consolidations; pleural effusions; and, often, dramatic hilar Microbiologic Tests


lymphadenopathy (Figure 43-4). Diagnostic assays available for Nocardia infection in dogs and
cats are shown in Table 43-2.
Sonographic Findings
Abdominal ultrasound findings in dogs and cats with dissemi- Isolation and Identification
nated nocardiosis have not been described in detail because dis- Nocardia spp. grow aerobically at a wide temperature range on
seminated disease with abdominal organ involvement is rare. simple media (e.g., Sabouraud’s glucose agar, blood agar). Organ-
However, intraparenchymal mass lesions and flocculent ascites isms are usually recovered in pure cultures, and colonies are often
fluid might be expected. visible after 2 days. However, in some cases, 2 to 4 weeks of
incubation may be necessary, and the organism may be over-
Advanced Imaging grown by contaminating bacteria. Thus, the laboratory should
Nodular lung lesions similar to those described in humans were be alerted if nocardiosis is suspected, so that selective media and
identified with computed tomography in one dog with dissemi- long incubation times are used. Colonies are smooth and moist,
nated nocardiosis.5 Computed tomographic and MRI abnor- or rugose with a powdery surface due to aerial filamentation, and
malities in dogs and cats with confirmed CNS nocardiosis have may be pigmented. Organisms may then be identified based on
not been reported. One dog with suspected nocardiosis based their microscopic appearance and their ability to take up acid fast
on histopathology had a large, T2-hyperintense circumscribed stains. However, not all pathogenic strains of Nocardia species
lesion in the occipital cortex that was consistent with a brain are acid fast.19,20,30 L-form Nocardia spp., which are cell wall–
abscess, together with widespread cerebral edema and hernia- deficient variants, have been associated with disease in people
tion of the cerebellum through the foramen magnum.4 and a dog.31 These bacteria require special media for isolation
and culture, similar to mycoplasmas (see Chapter 39).
Traditionally, Nocardia species have been distinguished based
on their growth characteristics and antibiotic susceptibility pat-
terns; however, molecular methods provide a more reliable and
rapid means of speciation. Sequence analysis of PCR products
from the 16S rRNA gene identifies most pathogenic Nocardia
spp., but differentiation of closely related species may require
analysis of other genes, multilocus sequence typing, or DNA-
DNA hybridization.2,17 DNA-DNA hybridization is considered
the gold standard for nocardial species determination.2 Matrix-
assisted laser desorption/ionization–time of flight (MALDI-
TOF) mass spectrometry shows promise for rapid and reliable
identification of Nocardia species in clinical microbiology lab-
oratories.32 PCR assays have also been used to directly detect
Nocardia spp. in clinical specimens, without prior isolation.2
Because Nocardia organisms are ubiquitous in soil and may
be inhaled by healthy animals, the isolation of small numbers
FIGURE 43-4  Lateral thoracic radiograph of a 4-year-old male neutered domestic of organisms from ulcerated skin lesions or the respiratory tract
shorthair cat with disseminated nocardiosis. Pleural effusion is present and obscures the may not be clinically significant and must be interpreted in con-
cardiac silhouette. A mass is associated with the caudal aspect of the left caudal lung junction with clinical signs and history of immune compromise.
lobe. Repeated positive cultures, pure cultures, or isolation of large

TABLE 43-2
Diagnostic Assays Available for Nocardia Infection in Dogs and Cats
Assay Specimen Type Target Performance
Culture Aspirates of purulent material Nocardia spp. Grows on most simple media. May require
(e.g., abscesses, pleural effusion); several weeks’ incubation. Because Nocardia
whole blood; tissue specimens spp. are soil saprophytes, isolation from skin
obtained by biopsy or at nec- lesions does not imply disease causation.
ropsy (e.g., subcutaneous tissue, PCR and sequencing may be required to
lung tissue) identify the species present.
Cytology or histo- Aspirates or tissue specimens Nocardia spp. Organisms are gram-positive, variably acid-
pathology with organisms fast, and are not typically accompanied
organism detection by sulfur granules, other bacteria, or the
using special stains Splendore-Hoeppli phenomenon. Definitive
diagnosis requires culture because non–acid-
fast Nocardia can resemble Actinomyces spp.
CHAPTER 43  Nocardiosis 413

numbers of the organism also suggest nocardiosis, and isola- or within abscesses. Although formation of sulfur granules by
tion of a single colony from a normally sterile site is significant. Nocardia spp. is uncommon compared with Actinomyces spp.,
Visualization of gram-positive filamentous bacteria in cytology small granules are occasionally found in skin lesions and pleural
specimens in association with acute inflammatory cells also and peritoneal fluid.10,22,28,33
assists in determination of clinical significance. Histopathology usually reveals a central region of necro-
The susceptibility of Nocardia spp. to antimicrobial drugs sis and suppuration, which, depending on the host immune
varies considerably between different Nocardia species, and so response, may be surrounded by macrophages, lymphocytes,
species identification can be used to guide antimicrobial drug and plasma cells (Figure 43-5, A). In chronic cutaneous-
selection (see Treatment, later) (Table 43-3). Susceptibility test- subcutaneous infections, pyogranulomatous foci may be inter-
ing for Nocardia spp. is difficult and should be done by labora- spersed within dense fibrous tissue. Plant material may be
tories with special expertise. Susceptibility testing may be helpful observed if plants were involved in inoculation of Nocardia
when there is a failure to respond to therapy or relapse occurs spp. into tissues, but this is more common with actinomycosis.
from drug resistance. It may also be useful if there are concerns Nocardia filaments are usually present in abundance within
that relate to adverse drug reactions to sulfonamides, the initial regions of necrosis and suppuration. They are poorly visible
treatment of choice (e.g., in dogs with immune-mediated disease in tissue sections stained with hematoxylin and eosin or with
that is complicated by cyclosporine-associated nocardiosis). Gridley’s fungal or periodic acid–Schiff. Organisms can be
Testing is also indicated when N. farcinica is present, which stained with Gram stain (see Figure 43-5, B) or methenamine
tends to be highly resistant to antimicrobial drugs, or when a silver preparations,5 especially with prolonged silver nitrate
nocardial species with unpredictable susceptibility is isolated. exposure. They are usually partially acid fast and have the same
appearance described previously in the Cytology section. In
Pathologic Findings chronic skin infections, tissue granules characterized by colo-
Nocardiosis is characterized by suppurative necrosis and abscess nies arranged in large, rosette-like arrays have been described.
formation and infrequently granulomas (see Figure 43-3). Gross In contrast to actinomycosis, other bacteria and the Splendore-
lesions in organs such as the spleen, lung, liver, and the brain Hoeppli phenomenon are not generally present.
typically consist of numerous small (1 mm) to large (several
centimeters), discrete or coalescing, intraparenchymal white or Treatment and Prognosis
gray-white nodules. On cut section, nodules appear caseous to
purulent. Lymph nodes are enlarged, often massively, and are Treatment
firm to fluctuant with a caseous to purulent core. A reddish- Successful treatment of nocardiosis relies on the combina-
brown exudate may be present in the pleural or peritoneal space tion of appropriate antimicrobial therapy combined with the

TABLE 43-3
Suggested Antimicrobial Drug Selection for Treatment of Pathogenic Nocardia spp. Based on Minimum Inhibitory
Concentration Data
Appropriate Antimicrobial
Nocardia Species Drug Choices Antimicrobial Drugs to Avoid
N. abscessus Ampicillin, amoxicillin–clavulanic acid, Imipenem, ciprofloxacin, clarithromycin
ceftriaxone, linezolid, amikacin and
gentamicin, sulfamethoxazole
N. brevicatena/paucivorans complex Ampicillin, amoxicillin–clavulanic acid, Gentamicin, clarithromycin
ceftriaxone, amikacin, sulfamethoxazole,
ciprofloxacin
N. nova complex (includes N. nova, Ampicillin, sulfamethoxazole, Amoxicillin–clavulanic acid
N. africana) ­erythromycin, clarithromycin,
­ceftriaxone, imipenem, amikacin
N. farcinica Amikacin, sulfamethoxazole, ciprofloxacin Ampicillin, broad-spectrum cephalospo-
(most isolates), imipenem (most isolates) rins, clarithromycin, aminoglycosides
other than amikacin
N. cyriacigeorgica Ceftriaxone, amikacin, imipenem Ampicillin, amoxicillin–clavulanic acid,
clarithromycin, ciprofloxacin
N. brasiliensis Minocycline, amoxicillin–clavulanic acid, Ampicillin, ciprofloxacin, clarithromycin
sulfamethoxazole
N. otitidiscaviarum Gentamicin, amikacin, sulfamethoxazole, All β-lactam antibiotics
ciprofloxacin

Data from Brown-Elliott BA, Brown JM, Conville PS, et al. Clinical and laboratory features of the Nocardia spp. based on current molecular tax-
onomy. Clin Microbiol Rev 2006;19:259-282.
414 SECTION 2  Bacterial Diseases

use of surgical drainage or debridement. Chronic, extensive and, if available, susceptibility test results.17 However, in  vitro
lesions may require surgical debridement or resection. Appro- susceptibilities do not always translate into a clinical response
priate antimicrobial therapy that includes surgical drainage is in vivo. Sulfonamides, including trimethoprim-sulfonamide com-
not always effective, possibly because of an inadequate host binations (TMS), are the first line antimicrobial drugs for treat-
immune response.8 Existing immunosuppressive drug treatment ment of nocardiosis (Table 43-4). Treatment durations from
should be discontinued or reduced, provided it does not cause 1 to 3 months are recommended in people with cutaneous infec-
relapse of a life-threatening underlying disease process. tions, up to 6 months for uncomplicated pulmonary infections,
The initial selection of an antimicrobial drug or drug combina- and 12 months or longer for systemic infections or infections in
tion should take into account the site of the infection, the host those who are immunocompromised.17 Clinical improvement is
immune status, the infecting Nocardia species (see Table 43-3), generally observed within 7 to 10 days of starting treatment. High
doses of trimethoprim-sulfas given for long periods to dogs (and
to a lesser extent, cats) can produce a variety of adverse effects
such as keratoconjunctivitis sicca and myelosuppression (see
Chapter 8). If adverse drug reactions occur, selection of drugs
other than TMS should ideally be based on susceptibility test
results. If these are not available, choices should be made based
on published susceptibility data for different Nocardia species (see
Table 43-3). Nocardia nova is susceptible to amoxicillin but
resistant to amoxicillin-clavulanic acid because of induction of
chromosomal β-lactamase by clavulanic acid.8,10 Substitution of
antimicrobial drugs within a drug class may not provide effec-
tive treatment; for example, although susceptible to amikacin,
N. farcinia isolates are resistant to gentamicin, and although sus-
ceptible to minocycline, N. brasiliensis isolates are mostly resis-
tant to doxycycline.34 Two- or three-drug combinations including
TMS, amikacin, and either ceftriaxone or imipenem have been
used, pending the results of susceptibility tests to treat severe
A nocardiosis in human patients.2 A combination of amikacin and
imipenem should be effective against all isolates. Combination
treatment with amikacin and TMS was suggested for cats with
N. farcinica infections, which generally require an aggressive,
combination drug treatment.8 After susceptibility testing, mono-
therapy with linezolid has consistently been used with success to
treat some refractory Nocardia infections in humans.35 Nocardia
isolates from dogs and cats have high minimum inhibitory con-
centrations for fluoroquinolones,36 which are not generally recom-
mended for treatment.
If CNS disease is present, either high doses of TMS given
parenterally, or drugs with excellent CNS penetration, which
include third-generation cephalosporins, carbapenems, or line-
zolid should be considered. The use of linezolid in veterinary
patients is controversial because it is an important drug for
resistant bacterial infections in humans (see Chapter 8).

B Prognosis
FIGURE 43-5  Histopathologic findings in nocardiosis. A, Severe, necrotizing In a review of 53 dogs with nocardiosis, 50% of the dogs died
pyogranulomatous inflammation is present with intralesional bacteria, which are barely and 39% were euthanized.24 Of 36 cats with nocardiosis, 16 were
visible. H&E stain. B, Gram staining revealed tangles of filamentous, branching gram- either euthanized or died.8 The high mortality rate may relate to
positive bacteria. Brown and Benn stain. underlying immunosuppressive disease or immunosuppressive

TABLE 43-4
Medications That Could Be Used to Treat Dogs and Cats with Nocardiosis *
Drug Dose (mg/kg) Route Interval (hours)
Trimethoprim-sulfamethoxazole 30 PO, IV 12
Amikacin† 10-14 (cat), 15-30 (dog) IV, IM, SC 24
Imipenem-­cilastatin† 2-5 IV 8

*Pending species identification with or without susceptibility testing.


†Amikacin and imipenem-cilastatin are generally used in combination with each other or with trimethoprim-sulfamethoxazole.
CHAPTER 43  Nocardiosis 415

drug treatment, delayed diagnosis, and inappropriate treatment. be carefully examined for foreign material and cleaned promptly.
With earlier diagnosis and more aggressive, multidrug therapy, mor- Disease in both dogs and cats may be prevented by avoidance
tality of nocardiosis in animals may decrease to the rate reported in of excessive immunosuppression with potent drugs such as
people. Only 20% of humans with primary infections died, whereas cyclosporine.
42% of patients with predisposing conditions and more than 50%
of patients with systemic or CNS nocardiosis died.19 Public Health Aspects
Immunity and Vaccination No cases of human nocardiosis acquired from direct con-
tact with an animal have been reported; however, several
Immunity to nocardiosis is dependent on intact CMI. There are cases of cutaneous nocardiosis transmitted to people by cat
no vaccines for nocardiosis. scratches have been documented.37-40 Humans with impaired
CMI (such as those receiving immunosuppressive drug ther-
Prevention apy or those with HIV infection) should wear gloves and
practice hand washing after handling pets with nocardiosis,
Prevention of nocardiosis in cats involves housing cats indoors or another individual in the household should treat these
and limiting their fighting or biting activity. Any wounds should animals.

CASE EXAMPLE cornea and anterior chamber were clear with no ocular
discharge. Fundoscopic examination showed focal areas of
tapetal hyperreflectivity and focal dark and dull lesions with
Signalment: “Bob,” a 6-year-old male neutered domestic peripheral hyperreflectivity.
shorthair cat from San Francisco, CA Musculoskeletal: BCS 7/9, with generalized weakness,
History: Bob, a renal transplant recipient, was evaluated recumbency and unwillingness/inability to walk.
for a 2-week history of decreased activity and progressive Respiratory: Tachypnea with a shallow respiratory pattern was
inappetence and lethargy. He was taken to a local identified. Decreased lung sounds were present ventrally.
emergency clinic where blood work showed only mild All Other Systems: No clinically significant abnormalities were
hyponatremia (130 mmol/L) and a white cell count of 19,000 present. The transplanted kidney could be palpated in the
cells/µL. No abnormalities were detected on abdominal right abdomen. The urinary bladder was small.
ultrasound examination. Bob was treated overnight with Neurologic Examination:
IV fluids and sent home with instructions to administer Mentation: Obtunded.
amoxicillin-clavulanic acid (12.5 mg/kg PO q12h). However, Gait/Posture: The cat had a tendency to lean to the right, but
the cat’s condition failed to improve, and so he was taken could not or would not walk more than a few steps.
to a local veterinary clinic 3 days later. Further laboratory Cranial Nerves: Menace responses were absent bilaterally.
testing showed hyponatremia, hypoalbuminemia, and mild Postural Reactions: Delayed placing reactions were identified
hyperbilirubinemia. Urinalysis showed a specific gravity of in all four limbs.
1.054, 3+ bilirubin, 1+ hemoprotein, 0-2 WBC/HPF, 6-10 RBC/ Spinal Reflexes and Panniculus Reflex: Normal.
HPF, and no bacteria or casts. Aerobic bacterial urine culture Neuroanatomic Location: Cerebrothalamic, possibly right
was negative. The following day Bob became tachypneic sided.
and pleural effusion was identified. In-house examination Laboratory Findings:
of the effusion fluid revealed cloudy fluid that contained CBC:
large numbers of erythrocytes, degenerate neutrophils, HCT 30.4% (30%-50%), MCV 46.5 fL (42-53 fL)
and macrophages. No bacteria were seen with Gram stain. MCHC 32.6 g/dL (30-33.5 g/dL)
Bob was then referred to the University of California, Davis, Reticulocytes 18,000 cells/µL (7000-60,000 cells/µL)
Veterinary Medical Teaching Hospital for further evaluation. WBC 22,860 cells/µL (4500-14,000 cells/µL)
Bob had received a renal transplant at 3.5 years of age and Neutrophils 19,660 cells/µL (2000-9000 cells/µL)
since then had been treated with prednisone (0.5 mg/kg PO Band neutrophils 2286 cells/µL
q12h) and cyclosporine (4 mg/kg PO q12h). A recent whole Lymphocytes 457 cells/µL (1000-7000 cells/µL)
blood trough cyclosporine concentration was 1100 ng/mL. Monocytes 229 cells/µL (50-600 cells/µL)
Physical Examination: Eosinophils 229 cells/µL (150-1100 cells/µL)
Body Weight: 5.1 kg Basophils 0 cells/µL (0-50 cells/µL).
General: Severely obtunded, T = 100.8°F (38.2°C), HR = 240 Platelets clumped but adequate. Neutrophils had slight toxic
beats/min, RR = 55 breaths/min, mild generalized icterus, change, and band neutrophils had marked toxic change.
mucous membranes pink to icteric, CRT = 1 s, hydrated. Serum Chemistry Profile:
Eyes, Ears, Nose, and Throat: The only clinically significant Sodium 136 mmol/L (151-158 mmol/L)
abnormalities related to the eyes. Anisocoria was present Potassium 3.5 mmol/L (3.6-4.9 mmol/L)
with the left pupil larger than the right. Direct and Chloride 98 mmol/L (113-121 mmol/L)
consensual pupillary light reflexes were present, and the Bicarbonate 18 mmol/L (15-21 mmol/L)

Continued
416 SECTION 2  Bacterial Diseases

Phosphorus 4.3 mg/dL (3.2-6.3 mg/dL) FIP, a systemic bacterial infection, or neoplasia. However,
Calcium 8.3 mg/dL (9.4-11.4 mg/dl) 10 hours after admission, Bob developed respiratory and
BUN 48 mg/dL (18-33 mg/dL) cardiac arrest. The owner elected euthanasia.
Creatinine 0.8 mg/dL (1.1-2.2 mg/dL) Necropsy Findings: Gross necropsy findings consisted of 120
Glucose 140 mg/dL (73-134 mg/dL) mL of flocculent, serosanguinous fluid within the thoracic
Total protein 4.8 g/dL (6.6-8.4 g/dL) cavity and multifocal to coalescing, variably sized, white to
Albumin 1.4 g/dL (1.9-3.9 g/dL) gray, firm to caseous nodules that largely effaced the caudal
Globulin 3.4 g/dL (2.9-5.3 g/dL) lung lobes and extended through the pleural surface, with
ALT 19 U/L (28-106 U/L) adhesions to the thoracic diaphragm and pleura (see Figure
AST 27 U/L (12-46 U/L) 43-3, A). A 3-mm nodule was also present in the wall of
ALP 5 U/L (14-71 U/L) the left ventricle. The peritoneal cavity contained multiple
Gamma GT 0 U/L (0-4 U/L) adhesions from the small intestines to the omentum and
Cholesterol 141 mg/dL (89-258 mg/dL) the stomach to the left abdominal wall. The right medial
Total bilirubin 1.5 mg/dL (0-0.2 mg/dL). liver lobe contained a focal, white to gray mass that was 10
Imaging Findings: cm in diameter and oozed purulent material when cut. The
Thoracic Radiographs: See Figure 43-4. There was a moderate spleen contained multiple, white, raised, pinpoint foci. The
amount of pleural effusion. A mass was associated with native kidneys were shrunken, pale, irregular, and had loss of
the caudal aspect of the left caudal lung lobe. The cardiac corticomedullary definition. The right transplanted kidney
silhouette was completely obscured on the lateral projection. contained 3-4, tan, firm, homogenous cortical nodules that
On the ventrodorsal projection, there appeared to be a mass ranged from 2 to 4 mm in diameter. On cut section, the brain
effect superimposed over the caudal border of the cardiac contained multifocal, randomly distributed, soft, white to
silhouette that deformed the left and right caudal lung lobes. gray, homogenous foci that measured 1-4 mm in diameter
Abdominal Ultrasound: The native kidneys were markedly (see Figure 43-3, B). Histopathology of the lung, pleural,
diminished in size, were irregular in contour, and myocardium, thoracic diaphragm, spleen, liver, transplanted
had a markedly irregular parenchyma with a lack of kidney, adrenal medulla, stomach, salivary gland, brain,
corticomedullary distinction. The transplanted kidney and meninges revealed chronic, severe, necrotizing
appeared within normal limits. The muscularis layer of the pyogranulomatous inflammation with intralesional
small intestine was mildly thickened diffusely. A Y-shaped branching filamentous bacteria. There was also chronic,
region of mineralization was identified within the right liver moderate, multifocal and necrotizing pyogranulomatous
lobes, which was likely associated with the biliary system. choroiditis and cyclitis in both eyes. The bacteria were gram
Treatment and Outcome: Thoracocentesis was performed, positive but were not acid fast (see Figure 43-5, B).
and 35 mL of turbid red fluid was removed. Unfortunately, Microbiologic Testing: Aerobic and anaerobic bacterial
the cat became progressively more agitated and tachypneic, culture (liver, lung specimens collected at necropsy): Small
thoracocentesis was aborted, and Bob was placed in a numbers of Nocardia nova. No other bacteria were cultured.
cage with oxygen supplementation. Preliminary cytologic Diagnosis: Disseminated Nocardia nova infection
examination of the fluid yielded results that were consistent Comments: In this case, nocardiosis followed a rapid
with referring veterinarian’s findings. Treatment with IV clinical course, as can occur in human patients with severe
crystalloid fluids (0.9% NaCl with 20 mEq/L KCl at 12 mL/ immunodeficiency. The trough whole blood cyclosporine
hr), enrofloxacin (2.5 mg/kg IV q12h), and ampicillin (22 mg/ concentration was high; the goal of treatment is to obtain
kg IV q8h) was initiated, and pleural fluid was submitted to blood concentrations of approximately 300 to 500 ng/
the laboratory for cytologic examination and aerobic and mL, although the optimum concentration that achieves
anaerobic bacterial culture. The plan was to stabilize Bob adequate but not excessive immunosuppression in cats is
with this treatment until additional thoracocentesis and not known.18 Treatment with antimicrobial drugs was not
chest tube placement could be performed. Differential effective given the advanced nature and severity of the
diagnoses considered were cryptococcosis, toxoplasmosis, disease in this cat.

SUGGESTED READINGS 2. Brown-Elliott BA, Brown JM, Conville PS, et al. Clinical and labo-
ratory features of the Nocardia spp. based on current molecular
Brown-Elliott BA, Brown JM, Conville PS, et al. Clinical and laboratory taxonomy. Clin Microbiol Rev. 2006;19:259-282.
features of the Nocardia spp. based on current molecular taxonomy. 3. Paul AE, Mansfield CS, Thompson M. Presumptive Nocardia spp.
Clin Microbiol Rev. 2006;19:259-282. infection in a dog treated with cyclosporin and ketoconazole. N Z
MacNeill AL, Steeil JC, Dossin O, et al. Disseminated nocardiosis caused Vet J. 2010;58:265-268.
by Nocardia abscessus in a dog. Vet Clin Pathol. 2010;39:381-385. 4. Smith PM, Haughland SP, Jeffery ND. Brain abscess in a dog
Malik R, Krockenberger MB, O’Brien CR, et al. Nocardia infections in immunosuppressed using cyclosporin. Vet J. 2007;173:675-678.
cats: a retrospective multi-institutional study of 17 cases. Aust Vet J. 5. MacNeill AL, Steeil JC, Dossin O, et  al. Disseminated nocar-
2006;84:235-245. diosis caused by Nocardia abscessus in a dog. Vet Clin Pathol.
2010;39:381-385.
REFERENCES 6. Ribeiro MG, Salerno T, Mattos-Guaraldi AL, et al. Nocardiosis: an
1. Nocard ME. Note sur la maladie des boeufs de la Guadeloupe con- overview and additional report of 28 cases in cattle and dogs. Rev
nue sous le nom de farcin. Ann Inst Pasteur (Paris). 1888;2:293-302. Inst Med Trop Sao Paulo. 2008;50:177-185.
CHAPTER 43  Nocardiosis 417

7. Saubolle MA, Sussland D. Nocardiosis: review of clinical and labo- 25. Lobetti RG, Collett MG, Leisewitz A. Acute fibrinopurulent pneu-
ratory experience. J Clin Microbiol. 2003;41:4497-4501. monia and haemoptysis associated with Nocardia asteroides in
8. Malik R, Krockenberger MB, O’Brien CR, et  al. Nocardia infec- three dogs. Vet Rec. 1993;133:480.
tions in cats: a retrospective multi-institutional study of 17 cases. 26. Ackerman N, Grain E, Castleman W. Canine nocardiosis. J Am
Aust Vet J. 2006;84:235-245. Anim Hosp Assoc. 1982;18:147-153.
9. Sykes JE. Unpublished observations. 2012. 27. Beaman BL, Sugar AM. Nocardia in naturally acquired and experi-
10. Hirsh DC, Jang SS. Antimicrobial susceptibility of Nocardia nova mental infections in animals. J Hyg (Lond). 1983;91:393-419.
isolated from five cats with nocardiosis. J Am Vet Med Assoc. 28. Tilgner SL, Anstey SI. Nocardial peritonitis in a cat. Aust Vet J.
1999;215:815-817, 795-816. 1996;74:430-432.
11. Hattori Y, Kano R, Kunitani Y, et al. Nocardia africana isolated 29. Mealey KL, Willard MD, Nagode LA, et al. Hypercalcemia associ-
from a feline mycetoma. J Clin Microbiol. 2003;41:908-910. ated with granulomatous disease in a cat. J Am Vet Med Assoc.
12. de Farias MR, Werner J, Ribeiro MG, et al. Uncommon mandibu- 1999;215:959-962, 946.
lar osteomyelitis in a cat caused by Nocardia africana. BMC Vet 30. Lerner PI. Nocardiosis. Clin Infect Dis. 1996;22:891-903; quiz
Res. 2012;8:239. 904-905.
13. Harada H, Endo Y, Sekiguchi M, et al. Cutaneous nocardiosis in a 31. Buchanan AM, Beaman BL, Pedersen NC, et al. Nocardia asteroi-
cat. J Vet Med Sci. 2009;71:785-787. des recovery from a dog with steroid- and antibiotic-unresponsive
14. Ajello L, Walker WW, Dungworth DL, et al. Isolation of Nocar- idiopathic polyarthritis. J Clin Microbiol. 1983;18:702-708.
dia brasiliensis from a cat with a review of its prevalence and geo- 32. Verroken A, Janssens M, Berhin C, et  al. Evaluation of matrix-
graphic distribution. J Am Vet Med Assoc. 1961;138:370-376. assisted laser desorption ionization–time of flight mass spec-
15. Ramos-Vara JA, Wu CC, Lin TL, et  al. Nocardia tenerifensis trometry for identification of Nocardia species. J Clin Microbiol.
genome identification in a cutaneous granuloma from a cat. J Vet 2010;48:4015-4021.
Diagn Invest. 2007;19:577-580. 33. Davenport DJ, Johnson GC. Cutaneous nocardiosis in a cat. J Am
16. Luque I, Astorga R, Tarradas C, et al. Nocardia otitidiscaviarum Vet Med Assoc. 1986;188:728-729.
infection in a cat. Vet Rec. 2002;151:488. 34. Gomez-Flores A, Welsh O, Said-Fernandez S, et  al. In  vitro and
17. Sorrell TC, Mitchell DH, Iredell JR, et  al. Nocardia species. In: in  vivo activities of antimicrobials against Nocardia brasiliensis.
Mandell GL, Bennett JE, Dolin R, eds. Principles and Practice of Antimicrob Agents Chemother. 2004;48:832-837.
Infectious Diseases. Philadelphia, PA: Elsevier; 2010:3199-3207. 35. Kobayashi N, Sueoka-Aragane N, Naganoby N, et  al. Dissemi-
18. Edwards DF. Actinomycosis and nocardiosis. In: Greene CE, ed. nated nocardiosis caused by Nocardia concava with acute respira-
Infectious Diseases of the Dog and Cat. 3rd ed. St Louis, MO: tory failure and central nervous system involvement treated with
Saunders Elsevier; 2006:451-461. linezolid. Intern Med. 2012;51:3281-3285.
19. Beaman BL, Beaman L. Nocardia species: host-parasite relation- 36. Govendir M, Norris JM, Hansen T, et al. Susceptibility of rapidly
ships. Clin Microbiol Rev. 1994;7:213-264. growing mycobacteria and Nocardia isolates from cats and dogs to pra-
20. McNeil MM, Brown JM. The medically important aerobic acti- dofloxacin. Vet Microbiol. 2011;153:240-245.
nomycetes: epidemiology and microbiology. Clin Microbiol Rev. 37. Astudillo L, Dahan S, Escourrou G, et al. Cat scratch responsible
1994;7:357-417. for primary cutaneous Nocardia asteroides in an immunocompetent
21. Kadar E, Sykes JE, Kass PH, et  al. Evaluation of the prevalence patient. Br J Dermatol. 2001;145:684-685.
of infections in cats after renal transplantation: 169 cases (1987- 38. Bottei E, Flaherty JP, Kaplan LJ, et al. Lymphocutaneous Nocardia
2003). J Am Vet Med Assoc. 2005;227:948-953. brasiliensis infection transmitted via a cat scratch: a second case. Clin
22. Campbell B, Scott DW. Successful management of nocardial Infect Dis. 1994;18:649-650.
empyema in a dog and cat. J Am Anim Hosp Assoc. 1975;11: 39. Freland C, Fur JL, Nemirovsky-Trebucq B, et al. Primary cutane-
769-773. ous nocardiosis caused by Nocardia otitidiscaviarum: two cases and a
23. Cross RF, Nagao WT, Morrison RH. Canine nocardiosis; a report review of the literature. J Trop Med Hyg. 1995;98:395-403.
of two cases. J Am Vet Med Assoc. 1953;123:535-536. 40. Sachs MK. Lymphocutaneous Nocardia brasiliensis infection
24. Marino DJ, Jaggy A. Nocardiosis. A literature review with selected acquired from a cat scratch: case report and review. Clin Infect Dis.
case reports in two dogs. J Vet Intern Med. 1993;7:4-11. 1992;15:710-711.
CHAPTER 44

Mycobacterial Infections
Jane E. Sykes and Danièlle A. Gunn-Moore

Mycobacterium tuberculosis Complex


Overview of Mycobacterial Infections in Dogs
and Cats Etiology and Epidemiology
First Described: Evidence of tuberculosis in humans dates
back to 2400 to 3400 bc, but the causative agent was not MTBC bacteria include the slowly growing, host-associated
demonstrated until 1882 (Robert Koch).1 bacteria Mycobacterium tuberculosis, Mycobacterium bovis,
Causes: Mycobacterium spp. (order Actinomycetales, family Mycobacterium microti, Mycobacterium africanum, Myco-
Mycobacteriaceae) bacterium pinnipedii, Mycobacterium caprae, and Mycobac-
terium canettii. These organisms cause tuberculosis in humans
Geographic Distribution: Worldwide, but the distribution of and animals. Only M. tuberculosis, M. bovis, and M. microti
different mycobacterial species varies. infect dogs and cats (Table 44-2). M. africanum and M. canettii
Major Clinical Signs: Cutaneous nodular, ulcerated, or drain- are rare causes of human tuberculosis in Africa, M. pinnipedii
ing skin lesions; peripheral or internal lymphadenopathy; infects seals, and M. caprae is primarily a ruminant pathogen.
pneumonia with cough and/or tachypnea; osteomyelitis; MTBC organisms share greater than 95% DNA-DNA homol-
granulomatous or pyogranulomatous infiltrates in a vari- ogy, and so are difficult to differentiate from one another, even
ety of abdominal organs and, rarely, the central nervous with many molecular methods. Unlike other mycobacterial spe-
system and eye. cies, MTBC bacteria do not survive well in the environment,
Differential Diagnoses: Neoplasia (especially lymphoma), although M. bovis can survive months in feces or carcasses.
feline infectious peritonitis (cats), tularemia, nocardiosis, Humans are the only reservoir host for M. tuberculosis, which
actinomycosis, rhodococcosis, bartonellosis, leishmani- is responsible for more than 90% of tuberculosis in humans.
asis, and fungal infections Cats are resistant to infection with M. tuberculosis. Disease due
to M. tuberculosis has been reported in dogs that reside in vari-
Human Health Significance: Transmission of mycobacteria ous parts of the United States, Africa, France, and Switzerland.2-9
from dogs or cats to humans has not been reported. How- Affected dogs from Europe have had travel histories; one was
ever, the potential for transmission of M. tuberculosis and from western Africa and the other was a stray from southern
M. bovis to humans exists. Humans acquire infection by Europe.2,3 Dogs are usually infected after prolonged aerosol
other mycobacteria from environmental sources. exposure to contaminated human respiratory secretions, so the
disease is a “reverse zoonosis” (or anthropozoonosis). Thus,
disease occurs in dogs where tuberculosis occurs in humans. In
the United States, M. tuberculosis infection is most prevalent in
humans from the northeast/mid-Atlantic states, southern states,

M
ycobacterium spp. are aerobic, nonmotile, non–spore- Alaska, California, and Nevada (Figure 44-1). Dogs have not
forming, gram-positive, acid-fast pleomorphic bacilli transmitted M. tuberculosis infection to humans despite pro-
that cause chronic infections in humans and animals. longed close contact in some situations,7 possibly because the
Mycobacterium spp. have a high cell wall mycolic acid content, pulmonary cavitary lesions that are required for transmission
which causes them to retain a pink color when stained with do not develop to the same extent in dogs as they do in humans.
acid-fast stains such as Ziehl-Neelsen or Kinyoun stains and However, organisms have been detected in dogs’ sputum, so
examined under the microscope. As a result, the term “acid-fast transmission remains possible.
bacilli” (AFB) is often associated with mycobacteria, although Cats are most often infected with M. microti or M. bovis.10
Nocardia is variably acid fast (see Chapter 43). Rodents (especially voles and shrews) are reservoir hosts for
Mycobacterium spp. belong to the family Mycobacteriaceae M. microti, and cattle and a variety of wildlife species are res-
and are grouped into those that belong to the Mycobacterium ervoir hosts for M. bovis. The overwhelming majority of feline
tuberculosis complex (MTBC), the Mycobacterium avium com- infections with M. microti and M. bovis have been reported from
plex (MAC), the lepromatous mycobacteria, and nontubercu- the United Kingdom, where M. microti infection is endemic in
lous mycobacteria other than M. avium (Table 44-1). Disease voles, wood mice, and shrews, and M. bovis infection is endemic
caused by these groups of microorganisms differs in its epide- in Eurasian badgers and cattle. Infections with M. microti and
miology, pathogenesis, clinical presentation, zoonotic potential, M. bovis comprise a third of feline infections with culturable
and response to antimicrobial treatment. Accordingly, each mycobacteria in the United Kingdom.10 Cats infected with
group is considered separately in this chapter. M. bovis tend to be young adult cats (median age 3 years),

418
CHAPTER 44  Mycobacterial Infections 419

TABLE 44-1
Mycobacterial Pathogens of Clinical Significance in Dogs and Cats
Group Mycobacterial Species Description
M. tuberculosis complex M. tuberculosis, M. bovis, M. microti Host-associated, slow-growing species. Cause cutaneous
and disseminated granulomatous disease in dogs (all three
species) and cats (M. bovis and M. microti)
M. avium-intracellulare M. avium subsp. avium, M. avium Opportunistic, environmental, slow-growing species. Cause
complex subsp. hominissuis cutaneous and disseminated granulomatous disease in
dogs (subsp. hominissuis) and cats (subsp. avium). Some
may cause leprosy syndromes in cats.
Lepromatous mycobacteria M. lepraemurium, the CLG organism, Highly fastidious or unculturable mycobacteria. Cause
and other unnamed species, some nodular granulomatous to pyogranulomatous cutaneous
with genetic resemblance to known disease in cats (all species) or dogs (CLG organism).
nontuberculous mycobacteria
Rapidly growing M. fortuitum, M. smegmatis, Opportunistic, environmental, rapidly growing species.
­nontuberculous M. abscessus, M. chelonae, Cause pyogranulomatous panniculitis, pneumonia, or dis-
­mycobacteria M. ­thermoresistibile, M. goodii, seminated infections in cats or dogs
M. flavescens, M. alvei
Slowly growing M. kansasii, M. ulcerans, Opportunistic, environmental, slowly growing species.
­nontuberculous M. ­genavense, M. malmoense, Cause pyogranulomatous cutaneous or disseminated
­mycobacteria other than M. celatum, M. terrae, M. simiae, disease in dogs or cats.
M. avium ‘M. visibile’*

CLG, Canine leproid granuloma.


*The exact position of ‘M. visibile’ is not clear; it has also been grouped within the lepromatous mycobacteria.

TABLE 44-2
Organisms of the Mycobacterium tuberculosis Complex of Clinical Significance in Dogs and Cats
Major Geographic Distribution
Reservoir of Reported Cases in Human Health
Species Host(s) Hosts Companion Animals Clinical Signs Significance
M. tuberculosis Dogs Humans United States, Africa, Pneumonia and tracheo­ Primary cause of tuber-
southern Europe bronchial lymphadenopa- culosis in humans.
thy; rarely dissemination Reverse zoonosis.
to the CNS, liver, kidney Transmission back to
humans not reported
but may be possible.
M. bovis Cats, rarely Cattle, Southwestern England and Cutaneous lesions, periph- Rare cause of tubercu-
dogs Eurasian Wales, New Zealand, eral lymphadenopathy, losis in humans but
badgers, Argentina, rarely occasionally dissemina- transmission from
other wildlife United States tion to abdominal lymph animals to humans
species nodes and other organs, theoretically possible
osteomyelitis
M. microti Cats, very Voles, wood Southwestern Scotland, Cutaneous lesions, periph- Very rare cause of tu-
rarely dogs mice, shrews, northern and southern eral lymphadenopathy, less berculosis in humans
camelids England, western Europe often pneumonia, arthritis,
osteomyelitis, ocular lesions

whereas the median age of cats infected with M. microti is M. bovis infections mainly occur in cats that reside in
8 years.10 All affected cats have been more than 1 year of age, Wales and southwestern England and that have access to the
which likely reflects the long incubation period associated with outdoors (Figure 44-2).10 Feline M. bovis infections also occur
these infections. Retroviral infection does not appear to predis- in New Zealand, where brushtail possums act as reservoir
pose to infection. hosts11; Argentina, where stray cats are often fed raw cattle
420 SECTION 2  Bacterial Diseases

FIGURE 44-1  Geographic distribution of human cases of Mycobacterium tuberculosis infection in the United States, 2005-2011. Compiled from Morbidity and Mortality Weekly reports
data on trends in human tuberculosis, years 2005, 2007, 2008, 2010, and 2011. *The intensity of the red color reflects the absolute number of cases/year, and whether case numbers were
persistently high or varied among the 5 years evaluated. Thus the states that persistently have the highest rates of M. tuberculosis infection are Alaska, Hawaii, California, and New York
(Long Island).

A study from the United States showed no evidence of trans-


mission of M. bovis to cats or dogs that lived on infected
cattle farms.15 Major wildlife reservoirs in North America,
which include white-tailed deer, bison, and elk, are less likely
than small mammals to interact with cats, which may explain
why fewer M. bovis infections are reported in cats from North
America when compared with the United Kingdom. Dogs are
uncommonly infected with M. bovis.16-18 Wounds inflicted
by badgers or a squirrel have preceded disease in affected
dogs.16 In recent years, disease in dogs has been restricted to
southwestern England or Ireland.
M. microti infections mainly occur in cats with outdoor
access from the United Kingdom, with major clusters in south-
western Scotland/northern England and southern England (see
Figure 44-2).10,19 Spillover of disease from voles to cats is sus-
pected.19 Vole and llama genotypes of M. microti have been
identified. Cats in the United Kingdom and one in Switzerland
have been infected with the vole type.20 A dog from France was
infected with the llama type.21

Clinical Features
Signs and Their Pathogenesis
MTBC organisms are inhaled (M. tuberculosis and possibly
M. bovis), ingested (M. bovis or M. microti), or inoculated
into the skin (M. bovis or M. microti) and replicate locally.
Organisms are then ingested by macrophages but survive and
replicate within these cells. Macrophage destruction leads to
FIGURE 44-2  Geographic distribution of Mycobacterium bovis and Mycobacte- recruitment of lymphocytes and additional monocytes, which
rium microti isolates from cats in the United Kingdom. (Redrawn from Gunn-Moore DA, initiates tubercle formation. Infected macrophages also travel
McFarland SE, Brewer JI, et  al. Mycobacterial disease in cats in Great Britain: I. Culture to local lymph nodes, with development of a primary complex
results, geographical distribution and clinical presentation of 339 cases. J Feline Med Surg lesion. If cell-mediated immunity (CMI) is defective, dissemi-
2011;13:934-944.) nation to distant sites can occur. Delayed type hypersensitiv-
ity (DTH) responses can control the initial infection or may
lead to central necrosis and calcification of the primary com-
lung12; and rarely the United States.13 In the United Kingdom, plex lesions, with persistence of organisms in the center of the
spillover of infection from cattle, badgers, and possibly small lesions.
rodents to cats is suspected. Aggressive interactions between Clinical signs in dogs with pulmonary tuberculosis due to
cats and infected badgers may also be a route of transmission, M. tuberculosis are slowly progressive. They consist of chronic,
especially as most cutaneous lesions occur at sites where fight productive, or nonproductive cough, as well as inappetence,
and bite wounds often occur.10,14 Ingestion of unpasteurized weight loss, and lethargy.4,7 Rarely, dissemination to nonpul-
milk by cats is no longer a common route of transmission. monary sites such as the central nervous system (CNS), liver,
CHAPTER 44  Mycobacterial Infections 421

A B
FIGURE 44-3  Ulcerative skin lesion due to Mycobacterium microti infection in a 6-year-old male neutered domestic shorthair. A, Before treatment. B, After 2 months of treatment with
a combination of marbofloxacin, azithromycin, and rifampin.

or kidney leads to neurologic signs, weight loss, vomiting, or


diarrhea.2,3,9 Involvement of the lungs, liver, kidneys, and/or
lymph nodes can occur in dogs infected with M. bovis.16,18,22
Isolated lung and tracheobronchial node involvement similar to
that often reported for M. tuberculosis was described in one dog
that was infected with M. bovis.18
Most (>75%) cats infected with M. bovis and M. microti
have had cutaneous nodular lesions with or without mandibu-
lar lymphadenopathy. Multifocal peripheral lymphadenopathy
may be present.23 Mesenteric lymphadenopathy, pneumonia,
ocular lesions, and arthritis occur in some cats infected with
M. microti.20,23-25 Dissemination may result in weight loss,
fever, and inappetence. Lung involvement may be associated
with tachypnea, dyspnea, or cough.18

Physical Examination Findings


Dogs with pulmonary involvement caused by M. tuberculo-
sis may show fever, increased lung sounds, and cough. Other
physical examination findings that relate to dissemination of
infection are present less often, such as thin body condition,
neurologic signs, or hepatomegaly. Abnormal findings on
examination of the respiratory tract and/or abdominal organs
may also be detected in dogs with disseminated M. bovis or
M. microti infection. FIGURE 44-4  Mandibular lymphadenopathy in a cat infected with Mycobacterium
The most common physical examination findings in cats microti.
(and to a lesser extent, dogs) that are infected with M. bovis or
M. microti are single or multiple firm cutaneous nodules, and
enlargement of one or more peripheral lymph nodes (Figures Diagnosis
44-3 and 44-4). Cutaneous nodules may be mobile or fixed to
underlying tissues (muscle and bone) and can ulcerate or drain Diagnosis of MTBC infection relies on suggestive history, clinical
purulent fluid. Skin lesions are most prevalent on the head but signs, and radiographic abnormalities, combined with the results
can also occur on the limbs and the trunk.20,23 Enlarged mes- of histopathology, molecular tests, and culture. When mycobacte-
enteric lymph nodes, spleen, and/or liver may be detected on rial infection is suspected, clinical specimens should be divided into
abdominal palpation. Involvement of internal lymph nodes is portions—one for histopathology (in formalin), one for mycobac-
more common with M. bovis infections (21% of cats) than with terial culture, one for routine cultures for other bacterial species,
M. microti infections (9% of cats).10 Lameness may be present and one for mycobacterial PCR (fresh or frozen). The laboratory
in cats with skeletal involvement. should be informed that mycobacterial infection is a possibility.
422 SECTION 2  Bacterial Diseases

A B
FIGURE 44-5  Lateral thoracic radiograph (A) and 7-mm collimated computed tomography (CT) image immediately cranial to the carina (B) from a 9-year-old female spayed golden
retriever with pulmonary tuberculosis. A, Consolidation of the right cranial lung lobe is present together with interstitial to alveolar infiltrates in the right middle lung lobe. B, On the
CT scan, the right cranial lung lobe is consolidated and markedly volume reduced. Air bronchograms are present centrally that extend dorsally toward a focal region of aerated lung. The
granular mineral opacity adjacent to the right margin of the trachea represents mineralized lymphadenopathy of the right tracheobronchial lymph nodes. (From Sykes JE, Cannon AB, Norris
AJ, et al. Mycobacterium tuberculosis complex infection in a dog. J Vet Intern Med 2007;21:1108-1112.)

Laboratory Abnormalities
Complete Blood Count, Serum Biochemical Tests, and Urinalysis
Laboratory abnormalities in dogs and cats infected with MTBC
organisms are nonspecific. Mild anemia of inflammatory disease
and neutrophilic leukocytosis, sometimes with a left shift, and
monocytosis are often present. The serum chemistry profile may
show hypoalbuminemia and hyperglobulinemia. Hypercalce-
mia of granulomatous disease can also occur in dogs or cats.10,17
Increased serum liver enzyme activities have been reported in
dogs with hepatic involvement.3

Diagnostic Imaging
Plain Radiography
Findings on plain thoracic radiography in dogs infected with
M. tuberculosis or M. bovis include tracheobronchial or medias-
tinal lymphadenopathy,3,4,7 interstitial to alveolar infiltrates,22
pleural effusion,3 and lobar consolidation (Figure 44-5, A).7,22
Similar abnormalities can occur in cats infected with M. microti
and M. bovis (Figure 44-6). Bronchial, alveolar, nodular inter-
stitial, and unstructured interstitial patterns predominate in
cats, and perihilar lymphadenopathy can be present.20,26 Calci-
fication of pulmonary lesions or lymph nodes may be evident in
dogs or cats. Hepatomegaly, splenomegaly, or lymphadenopa-
thy, with or without ascites, may be present on abdominal radi-
ography, or abdominal radiographs may be unremarkable.26
Radiographic evidence of osteomyelitis (especially osteolysis)
can occur in cats infected with M. microti or M. bovis, often in FIGURE 44-6  Pneumonia in a 6-year-old male neutered oriental shorthair with sus-
association with overlying cutaneous lesions.26 pected Mycobacterium microti infection. Bronchoalveolar lavage revealed pyogranuloma-
tous inflammation, but acid-fast stains were negative. Results of an IFN-γ assay suggested
infection by M. microti.
Computed Tomography
Thoracic CT findings have been reported for a dog infected
with M. tuberculosis.7 Lung lobe consolidation, multifo-
cal stellate opacities, bronchial stenosis, bronchiectasis,
CHAPTER 44  Mycobacterial Infections 423

TABLE 44-3
Assays Available for Diagnosis of Mycobacterial Infections in Dogs and Cats
Assay Specimen Type Target Performance
Acid-fast staining Tissue aspirates, buffy Acid-fast bacteria Insensitive, especially for some mycobacterial
coat smears, body fluids, ­species such as MTBC bacteria. Other bacteria,
airway lavage ­specimens, such as Rhodococcus and Nocardia, may also be
tissues obtained at acid fast, so positive results are not specific for
­biopsy or necropsy ­Mycobacterium spp.
Gamma-interferon Whole blood (­peripheral Cell-mediated Not yet well validated, but show promise for
response assay blood mononuclear ­immune response discrimination between M. bovis and M. ­microti
cells) to ­mycobacterial infections in the United Kingdom. Variable
antigens ­responses to M. avium.
Mycobacterial See acid-fast staining; Culturable Some mycobacterial species are unculturable. Growth
culture avoid use of aspirates or ­Mycobacterium spp. of MTBC organisms and slowly growing nontuber-
swabs if possible culous mycobacteria may take weeks to months,
and identification requires special expertise. Allows
detailed molecular typing and susceptibility testing.
PCR See acid-fast staining Mycobacterial DNA Permits rapid identification of mycobacterial infection
and differentiates MTBC from other mycobacteria,
so appropriate precautions can be instituted. False-
negative results can occur with insufficient specimen
size, if there are very few bacteria present, or if
degradation of DNA occurs. False-positive results
are possible as a result of laboratory contamination.

tracheobronchial wall thickening, and partially mineralized Microbiologic Tests


masses consistent with enlarged mediastinal and tracheobron- Assays available for diagnosis of mycobacterial infections are
chial lymph nodes were present (Figure 44-5, B). In this dog, listed in Table 44-3.
the mineralization was not visible on plain thoracic radio-
graphs. CT in human patients with tuberculosis is considered Acid-Fast Stains
more sensitive than plain radiography for detection of pulmo- Acid-fast stains can be applied to smears or tissue sections for
nary lesions and enlarged lymph nodes and provides better detection of tuberculous mycobacteria. Organisms are faintly
visualization of calcifications. beaded bacilli, and are often within macrophages. The presence
of AFB suggests mycobacterial infection, but is not specific for
Sonographic Findings infection with MTBC organisms. Acid-fast staining of smears or
Abdominal sonography in animals with MTBC infections Cytospin preparations can be insensitive for detection of MTBC
may be unremarkable or reveal hepatomegaly, splenomegaly, organisms, and negative results do not rule out infection.
abdominal lymphadenopathy, and/or ascites. A dog with dis-
seminated M. tuberculosis infection had multifocal hypoechoic Immunologic Assays
masses within the kidneys and liver and mineralization of the Intradermal inoculation of tuberculin (a purified protein deriva-
hepatic and renal parenchyma.4 tive from M. tuberculosis), or the Mantoux test, is well vali-
dated and widely used in human medicine for diagnosis of, and
Cytologic Examination screening for, infection with MTBC organisms. Infected individ-
Cytologic examination of aspirates, effusion, or lavage uals develop a cutaneous DTH reaction at the site of inoculation
specimens from the respiratory tract of dogs with pulmo- 48 to 72 hours later. False-negative test results can occur as a
nary tuberculosis typically reveals a mixed population of result of underlying immunosuppression. False-positive results
often heavily vacuolated histiocytes and smaller numbers occur as a result of nontuberculous mycobacterial infection.
of degenerate or nondegenerate neutrophils and small lym- Cats do not respond reliably to tuberculin skin testing. In dogs,
phocytes.3,7,22 Intracytoplasmic, nonstaining bacilli were tuberculin testing has been performed on the medial surface of
seen in macrophages in a bronchoalveolar lavage specimen the pinna, but false-positive results have been documented with
from a dog infected with M. bovis.22 Moderate anisocytosis a variety of other bacterial infections. Tests for anti-mycobacte-
or anisokaryosis of exfoliated respiratory epithelial cells can rial antibodies are also not specific.27 Cell-based assays (ELISA
be present, which might cause suspicion for pulmonary neo- and ELISPOT), which measure IFN-γ production by patient
plasia.7 The presence of cholesterol crystals, caseous debris, peripheral blood mononuclear cells in response to specific myco-
and concentrically ­laminated crystalline structures known as bacterial antigens, have been used for diagnosis of tuberculosis
calcospherite bodies can be a clue for the presence of MTBC in humans and cattle. These assays have been adapted for use in
infection.7,22 cats and have the potential to rapidly distinguish infection with
424 SECTION 2  Bacterial Diseases

M. bovis from that with M. microti when responses to different result with a genus-specific assay but a negative result with an
mycobacterial antigens are compared.28 assay for MOTT organisms implies the presence of an MTBC
organism. The results of mycobacterial PCR assays should be
Bacterial Isolation and Identification interpreted in light of clinical findings, and PCR assays should
Routine aerobic, anaerobic, fungal, and mycoplasma cultures always be used with mycobacterial culture, which is the only
are negative when performed on specimens collected from ani- way to confirm the mycobacterial species involved and allows
mals with MTBC infections. Mycobacterial culture is required antimicrobial drug susceptibility testing. The latter is important
and is the gold standard for diagnosis of MTBC infection. The given the public health implications of MTBC infections.
use of laboratories with expertise in isolation of MTBC is rec-
ommended. Respiratory specimens such as respiratory lavages Pathologic Findings
are usually treated with the mucolytic N-acetylcysteine and a Gross Pathologic Findings
sodium hydroxide solution to destroy contaminating bacteria. The most frequent gross pathologic findings in dogs infected
Specimens from normally sterile sites, such as pleural fluid, are with M. tuberculosis are pulmonary congestion, lobar consoli-
not decontaminated so that viable mycobacteria are preserved dation, or granulomatous mass formation and sometimes mas-
as much as possible. Specimens are then inoculated onto spe- sive enlargement of tracheobronchial and mediastinal nodes.
cial liquid or solid media for isolation of mycobacteria, such as Lymph nodes may contain multifocal gritty, yellow, caseous
7H11 agar or Lowenstein-Jensen media (an egg-based medium). material.4,7,22 Pleural effusion may also be present.3 In dogs
Modifications to the media composition may be required on the with disseminated disease, pale nodular lesions may be found
basis of the mycobacterial species suspected, so good commu- in abdominal organs such the liver, peritoneal surfaces, and kid-
nication between the clinician and the microbiology laboratory neys.2,4,22 In one unusual case, a dog from Africa had nodular
can optimize the chance of successful culture. Growth is evident lesions that were confined to the liver and brain.2 Dogs infected
1 to 3 weeks after inoculation of liquid media, and 3 to 8 weeks with M. bovis have abnormalities similar to those infected
after inoculation of solid media. However, inoculation of solid with M. tuberculosis.16-18 In addition to cutaneous lesions and
media is still required, because some strains grow only on solid peripheral lymphadenomegaly, cats with disseminated M. bovis
media, and growth on solid media allows assessment of colony or M. microti infections may have abdominal lymphadeno-
morphology. Once growth is evident, nucleic acid–based assays, megaly and multifocal tumor-like, grayish-white masses within
mycolic acid analysis with high-performance liquid chromatog- parenchymal organs that can have hemorrhagic margins and/or
raphy, or mass spectrometry (MALDI-TOF)29 can be used to a soft purulent center. Pulmonary lesions are often grayish red
determine whether the organism belongs to the MTBC. Antimi- and may be accompanied by serosanguineous pleural effusion.
crobial drug susceptibility testing can also be performed.
Currently, performance of a series of phenotypic (biochemi- Histopathologic Findings
cal) assays on cultured isolates is the gold standard method Histopathologic findings are similar for all MTBC diseases.
for differentiation of mycobacterial species that belong to the Characteristic tubercles or granulomas are present in tissues,
MTBC. Molecular techniques such as PCR-restriction endonu- which vary in size and may coalesce (Figure 44-7). Tubercles
clease analysis, DNA probing, and 16S rRNA gene sequencing consist of abundant epithelioid macrophages and lesser num-
do not differentiate among MTBC species. Specialized molec- bers of neutrophils. Multinucleated giant cells are rarely present
ular techniques for further typing of MTBC isolates include in dogs or cats.4,30 The inflammatory foci are surrounded by
mycobacterial interspersed repetitive unit-variable-number a layer of macrophages, neutrophils, lymphocytes, and plasma
tandem repeat (MIRU-VNTR) typing and spoligotyping. Spo- cells and an outer layer (or background) of granulation tissue.
ligotyping is a PCR-hybridization typing method that targets The center of the tubercle may be necrotic. Central mineraliza-
the direct repeat region of MTBC strains. This region contains tion and lipid accumulation occurs in some dogs infected with
multiple DNA sequence repeats interspersed with nonrepetitive M. tuberculosis,7,22 but mineralization is rare in cats infected
spacer sequences. MTBC species vary in the numbers of repeats with M. microti or M. bovis. Acid-fast stains reveal low num-
and the presence or absence of some spacers and can be dif- bers of AFB in tubercle centers in most cases, but a careful
ferentiated on the basis of their hybridization patterns. Spoli- search may be required (Figure 44-8).4,7
gotyping also allows identification of prevalent M. tuberculosis
genotypes, such as the Beijing genotype, an aggressive, highly Treatment and Prognosis
transmissible strain that has been associated with multiple drug
resistance. MIRU-VNTR is often combined with spoligotyping Dogs and cats with suspected MTBC infection should be placed
for identification of MTBC organisms. Spoligotyping has been in isolation, and aerosol precautions are indicated (see Chapter
performed on several MTBC isolates from dogs and cats. 11). Treatment of dogs and cats with MTBC infection is contro-
versial because of concerns that relate to zoonotic potential. It
Molecular Diagnosis Using the Polymerase Chain Reaction should only be performed after owners have been fully educated
Real-time PCR-based assays are available on a commercial basis about the potential risks of disease transmission. Most affected
in some countries for rapid detection of Mycobacterium spp. dogs have been euthanized without specific treatment, but occa-
DNA in clinical specimens from dogs and cats. An understand- sionally euthanasia of animals with non-MTBC mycobacterial
ing of the range of mycobacterial species detected by an assay infection has occurred before a diagnosis of MTBC was con-
is of paramount importance for proper interpretation of the firmed. Combination drug therapy with the antimycobacterial
results. Genus-specific Mycobacterium spp. assays are available, drugs ethambutol, isoniazid, rifampin, and pyrazinamide is used
as well as assays that detect mycobacteria other than tuberculo- to treat human patients. Ethambutol and isoniazid interfere with
sis (MOTT) organisms. These assays can be used in combination mycobacterial cell wall synthesis; the mechanism of action of
to determine whether an MTBC organism is present. A positive pyrazinamide is not known, and M. bovis is naturally resistant
CHAPTER 44  Mycobacterial Infections 425

A B
FIGURE 44-7  Multifocal granuloma formation in the lung (A) and thoracic lymph node (B) of a 9-year-old female spayed golden retriever with pulmonary tuberculosis. The granu-
loma within the lymph node contains a lake of lipid and necrotic cellular debris, surrounded by an internal layer of epithelioid macrophages, a thick layer of fibrosis, and an external layer of
lymphocytes, plasma cells, and histiocytes. Fite and Ziehl-Neelsen acid-fast stains revealed scant numbers of acid-fast positive bacteria in areas of central necrosis (not shown).

a fluoroquinolone), followed by a continuation period with two


drugs (usually the macrolide and the fluoroquinolone, because
rifamycins are more often associated with adverse effects).
Esophagostomy tube placement may be necessary to facilitate
treatment and reduce the chance of exposure of the owner to the
infection through bite or scratch wounds.

Immunity and Vaccination


There are no vaccines available for prevention of mycobacterial
disease in animals.

Prevention
Prevention of M. tuberculosis infections in dogs relies on appro-
priate control and treatment of M. tuberculosis infections in
affected humans that are in close contact with dogs. Prophylaxis
with a single antimycobacterial drug such as isoniazid could be
considered for dogs that reside with affected humans, but iso-
niazid has the potential to cause adverse effects such as seizures
in dogs. The possibility of chronic infection by potentially zoo-
notic pathogens such as M. tuberculosis should be considered
FIGURE 44-8  Rare acid-fast bacteria in a biopsy specimen from a cat infected with
before stray dogs are adopted from underdeveloped countries.
M. microti.
In cats, prevention of M. bovis and M. microti infections can
be achieved when cats are housed indoors and rodent popula-
to it. Treatment of a dog with M. tuberculosis infection was tions controlled. The feeding of raw cattle or game (e.g., deer,
attempted with rifampin, isoniazid, and clarithromycin, but the elk) also has the potential to transmit M. bovis to dogs or cats
dog was euthanized after seizures developed.7 The seizures were and should be discouraged.
suspected to be an adverse effect of isoniazid administration.
Successful treatment of cats with cutaneous and/or pulmo- Public Health Aspects
nary MTBC infections has been achieved with combinations of
fluoroquinolones, clarithromycin or azithromycin, doxycycline, Disease caused by MTBC bacteria is usually notifiable to pub-
and a rifamycin; most successfully treated cases have received lic health authorities. More than 90% of human tuberculosis
a fluoroquinolone, clarithromycin or azithromycin, and a rifa- is caused by M. tuberculosis. Only 1% of cases result from
mycin (Table 44-4). Surgical excision of small skin lesions can M. bovis infection, and human infection with M. microti is very
be curative if dissemination is not present, but lesions may rare.31 Factors such as HIV infection, homelessness, and intra-
dehisce and progress if resection is not complete, and it may venous drug use predispose humans to M. tuberculosis infec-
not always be possible to determine whether dissemination has tion. Tuberculosis continues to be an important public health
occurred. The use of three drugs has been recommended for the problem worldwide and especially in Africa. It is second only
first 2 months of treatment (e.g., rifampin, clarithromycin, and to HIV as a cause of death worldwide from a single infectious
426 SECTION 2  Bacterial Diseases

TABLE 44-4
Suggested Drug Doses for Treatment of Mycobacterial Infections*
Interval
Drug Dose (mg/kg) Route (hours) Major Adverse Effects in Dogs and Cats
Isoniazid 5-10 PO 24 Hepatotoxicity, seizures. Start at a low dose; do not exceed
300 mg/day. Monitor liver enzymes.
Ethambutol 10-25 PO 48-72 Unknown; neuropathies, especially optic neuritis with visual
disturbances occur in humans.
Rifampin 5-10 PO 12-24 Gastrointestinal, hepatotoxicity, orange discoloration of urine,
tears, sclera, mucous membranes, feces. Monitor liver enzymes.
Clarithromycin 7.5 PO 12 Rare; hepatotoxicity and gastrointestinal signs possible.
Doxycycline 10 PO 12 Gastrointestinal
Enrofloxacin 5-10 (dogs only)† PO 24 Gastrointestinal
Marbofloxacin 2.75-5.5 PO 24 Gastrointestinal
Clofazimine 25 mg/cat, 4-8 mg/kg PO 24 Cutaneous photosensitization,‡ orange pigmentation to the skin,
(dogs) gastrointestinal. May be difficult to obtain in some countries.

*See text for recommended drug combinations for each mycobacterial species and additional drugs that may be useful.
†The use of other fluoroquinolones such as marbofloxacin or pradofloxacin is preferred in cats because of the risk of retinal toxicity (see Chapter 8).
‡Bennett S. Photosensitisation induced by clofazimine in a cat. Aust Vet J 2007;85:375-380.

agent,32 and drug resistance is emerging globally. Over the past an affected cat.36 The DNA of M. avium subsp. paratuberculo-
2 decades, case numbers have decreased to the lowest levels sis was detected in intestinal biopsies from dogs with chronic
in history in the United States.33 In the United States, the inci- gastrointestinal signs, but the role of this organism in disease
dence rate is highest among non-Hispanic Asians, non-Hispanic was not clear.37 Unlike MTBC organisms, which are associ-
blacks, and Hispanics, which is 25, 8, and 7 times greater, ated with reservoir hosts, MAC organisms are environmental
respectively, than the rate for non-Hispanic whites.33 Disease saprophytes with a worldwide distribution. They can be found
in immigrants to the United States results from reactivation of in soil, dust, and aquatic environments that include showers,
disease acquired in other countries. faucets, household drinking water, swimming pools, and hot
Public health authorities should be notified if a diagnosis tub spas.38 The organisms also infect free-living amoebae and
of MTBC infection is made in a dog or cat. An investigation form biofilms, which may promote environmental persistence.
may be performed by these authorities in order to identify the In the United Kingdom, a spatial cluster of infection has been
source of infection, and it may be necessary to perform one identified in cats from eastern England.10 Subclinical infections
or more Mantoux tests on individuals with significant contact with M. avium subsp. avium are common among birds, and
with infected animals. Although transmission of M. tuberculo- M. avium subsp. avium is the main cause of avian ­tuberculosis.39
sis from infected dogs to humans has not been reported as a Ingestion of infected birds or bird feces may be a route of trans-
result of direct contact, veterinary staff were infected when a mission to cats or dogs, but this requires clarification.
necropsy of an infected dog was performed, possibly following MAC organisms cause opportunistic infections in immu-
aerosolization of infected brain tissue when the skull was sec- nocompromised individuals. Most affected dogs and cats are
tioned with an electric saw.2 Properly fitted high-density surgi- 1 to 5 years of age. In dogs, disease occurs sporadically in any
cal masks (N95 masks) should be worn if surgery or necropsy is breed, but basset hounds, miniature schnauzers, and possibly
performed on a dog or cat suspected to have tuberculosis. Yorkshire terriers may be predisposed, possibly because of an
inherited CMI deficiency. In cats, retrovirus infection or treat-
Mycobacterium avium Complex ment with potent immunosuppressive drugs may predispose to
MAC infection, although most cats have no identifiable immu-
Etiology and Epidemiology nodeficiency. Siamese cats as well as Abyssinians and Somalis
may be predisposed.40,41 Disseminated MAC infection has also
The Mycobacterium avium complex (MAC) consists of two been reported in feline renal transplant recipients42,43 and in
closely related species, M. avium and Mycobacterium intra- a cat treated with chemotherapy for lymphoma.44 Infection is
cellulare. There are four subspecies of M. avium: hominissuis, not transmissible from one infected individual to another, but
avium, paratuberculosis, and silvaticum. M. avium subsp. disease may initially be indistinguishable from that caused by
hominissuis causes most human infections. The distribution MTBC bacteria. In the United States, although still rare, infec-
of subspecies in affected dogs and cats is currently not clear, tion of cats and dogs with MAC organisms is more common
because molecular methods are required for identification than infection with MTBC organisms. In the United Kingdom,
at the subspecies level. M. avium subsp. hominissuis infec- M. avium accounted for only 15% of 159 culturable mycobac-
tions have been identified in several dogs from continental terial infections in cats (73% were MTBC infections).10 The
Europe,34,35 whereas M. avium subsp. avium was isolated from median age of affected cats is 8 years.
CHAPTER 44  Mycobacterial Infections 427

FIGURE 44-9  Five-year-old female spayed husky dog with Mycobacterium avium
lymphadenitis. Swellings in the mandibular region are apparent.
FIGURE 44-10  Digital osteomyelitis in an 8-year-old female spayed Yorkshire terrier
dog with Mycobacterium avium infection. The first and second phalanges of the fifth digit
Clinical Features are destroyed, and there is irregular mineralization within a large soft tissue swelling in
this region. There is also moderate soft tissue swelling at the second digit. The third and
Signs and Their Pathogenesis fourth digits had been previously amputated at the mid-metacarpal bones, which are now
fused at their distal aspect.
Disease due to MAC organisms occurs when organisms in
soil, water, or dust are inhaled, ingested, or inoculated into
skin wounds in the face of host immunocompromise. Dis- lymphadenopathy, renomegaly, hepatomegaly, tachypnea, and/
seminated infection often leads to nonspecific signs such as or increased lung sounds. Rarely, neurologic signs such as nys-
fever, lethargy, inappetence, and weight loss, together with tagmus occur.41 In dogs, low-grade fever, a thin body condition,
signs that relate to specific organ involvement. In cats, dis- dehydration, mucosal pallor, tonsillar enlargement, lymphad-
ease resembles that caused by M. bovis or M. microti, with enomegaly (especially of the mandibular or abdominal nodes),
cutaneous lesions (especially on the head and limbs) in some hepatosplenomegaly, and/or abdominal pain may be found.
cats, osteomyelitis, pulmonary involvement with tachypnea Melena or hematochezia may be detected on rectal examina-
or cough, peripheral and abdominal lymphadenomegaly, and tion. Osteomyelitis may be associated with lameness or mass
gastrointestinal, liver, splenic, renal, omental, and uncom- lesions in association with underlying bone involvement (Fig-
monly CNS or marrow involvement.41,42,44-50 Gastrointesti- ure 44-10). A dog with spinal cord involvement had pelvic limb
nal involvement may initially manifest as weight loss despite paresis.53
a good appetite.41
Dogs with MAC infections often develop marked peripheral Diagnosis
or abdominal lymphadenopathy, tonsillar enlargement, hepato-
splenomegaly, and/or osteomyelitis. In some dogs, mandibular Laboratory Abnormalities
or cervical lymphadenopathy may be so severe that respiratory Complete Blood Count, Serum Biochemical Tests, and Urinalysis
distress and dysphagia occur (Figure 44-9).51 At necropsy, AFB Findings on routine laboratory tests in dogs and cats with MAC
and associated inflammatory lesions may also be found in the infections are similar to those described for MTBC infection.
small and large intestines, liver, spleen, lung and pleura, bone Most affected dogs or cats have had mild to moderate nonre-
marrow, and rarely the spinal cord.34,52-56 Involvement of the generative anemia, neutrophilia with bandemia, lymphopenia,
gastrointestinal tract may lead to chronic vomiting, diarrhea, hypoalbuminemia, hyperglobulinemia or hypoglobulinemia,
hematochezia, melena, inappetence, and weight loss. Intesti- and sometimes hypercalcemia, hyperbilirubinemia, and
nal rupture in one dog led to bacterial peritonitis. Even though increased liver enzyme activities.48,54,57 An absence of hemato-
inflammatory lesions with intralesional AFB have been detected logic or biochemical abnormalities has also been described.
in the lung at necropsy, pulmonary involvement is not a promi-
nent clinical feature of MAC infections in dogs, which may help Diagnostic Imaging
to distinguish the disease from M. tuberculosis infection. Plain Radiography
When radiographic abnormalities are present in cats infected
Physical Examination Findings with M. avium, they often consist of diffuse interstitial or
Physical examination of cats with MAC infections may reveal bronchointerstitial pulmonary patterns.41,48 Diffuse interstitial
fever, a thin body condition, cutaneous lesions similar to patterns may be present in the absence of clinical signs of respi-
those described for MTBC infection, peripheral or abdominal ratory involvement. Thoracic lymphadenopathy or mediastinal
428 SECTION 2  Bacterial Diseases

analysis. Subspecies determination requires specialized typing


techniques such as IS1245 restriction fragment length poly-
morphism typing, MIRU-VNTR, or the use of PCR assays that
detect gene fragments present in some M. avium subspecies but
not others.34,36,59

Molecular Diagnosis Using the Polymerase Chain Reaction


See the previous discussion of MTBC infection for information
on mycobacterial PCR assays.

Pathologic Findings
In general, pathologic findings in cats or dogs with MAC infec-
tion consist of focal, multifocal, or generalized lymphadeno-
megaly (up to 5 cm in diameter), and nodules in the spleen,
liver, lungs, omentum, intestinal wall, and occasionally the kid-
neys. Nodules are white, gray, or yellow and may be caseous
on cut section. Gastrointestinal mucosal hemorrhage may be
evident in dogs.54
FIGURE 44-11  Aspirate cytology from a cutaneous mass in a cat that developed Histopathology in MAC infections typically reveals large
after prolonged (>1 year) treatment with cyclosporine. Large numbers of negatively numbers of histiocytes and lesser numbers of neutrophils, which
stained mycobacterial organisms are present. (Image courtesy Dr. William Vernau, Univer- in some cases surround necrotic foci. Multinucleated giant cells
sity of California, Davis Veterinary Clinical Pathology Service.) are rarely seen, and mineralization has not been described.
Moderate to large numbers of AFB are usually present within
histiocytes (Figure 44-12).
masses may be present in dogs.35 Abdominal radiographs may
show abdominal masses or hepatosplenomegaly in both dogs Treatment and Prognosis
and cats. Radiographs of affected bone may reveal soft tissue
swelling and bony lytic lesions or periosteal proliferation (see Treatment of MAC infections in dogs or cats is challenging,
Figure 44-10). but apparent cure has been reported in a few cats.36,41,48 As
for infection with MTBC organisms, the use of two or three
Sonographic Findings drugs in combination is recommended to prevent emergence of
Abdominal sonography in animals with MAC infections resistance. The availability of macrolides (especially clarithro-
may be unremarkable or reveal hepatomegaly, splenomegaly, mycin) has greatly improved outcome for human patients with
hypoechoic nodules within the liver or spleen, intestinal wall MAC infections, so inclusion of clarithromycin or azithromycin
thickening, hyperechoic mesentery, or enlarged, hypoechoic, or is important. The currently recommended treatment for humans
heteroechoic abdominal nodes.58 with MAC infections consists of a ­rifamycin, ethambutol, and
a macrolide such as clarithromycin. Treatment of a dog with
Cytologic Examination a combination of drugs that did not include a macrolide was
Cytologic examination of aspirates from lymph nodes, abdomi- unrewarding.51 Various ­combinations of clarithromycin, clo-
nal organs, or respiratory lavage specimens from cats or dogs fazimine, doxycycline, rifamycins, and fluoroquinolones have
with M. avium infection reveals large numbers of histiocytes been used to treat affected cats (see Table 44-4). Clofazimine
with nonstaining intracellular bacteria (Figure 44-11) and lower is an ­antimycobacterial drug with an unknown mechanism of
numbers of neutrophils. action. In human patients, it has primarily been used to treat
lepromatous or rapidly growing opportunistic mycobacteria,
Microbiologic Tests but may have some activity against MAC organisms. It may be
For assays available for diagnosis of mycobacterial infections in difficult or impossible to obtain in some countries.
dogs and cats, see Table 44-3.
Public Health Aspects
Acid-Fast Stains
Application of acid-fast stains to smears or tissue sections can be Like dogs and cats, humans acquire MAC infections from the
used to identify MAC organisms in clinical specimens. In gen- environment; direct transmission from animals to people has
eral, AFB are identified more readily in MAC infections when not been described. Ownership of caged birds by immunocom-
compared with MTBC infections, and most reports of affected promised humans has been discouraged in the past because of
dogs and cats report the detection of organisms in cytology the potential that these birds might shed MAC organisms. How-
specimens stained with acid-fast stains. ever, human disease is caused by M. avium subsp. h ­ ominissuis
and not the avian organism (subsp. avium), so the feces of
Bacterial Isolation and Identification caged birds may not represent a significant source of infection.
See the previous discussion of MTBC infection for informa- Three clinical forms of MAC disease occur in people: local-
tion on mycobacterial culture. Like MTBC organisms, MAC ized pulmonary disease (which includes “hot tub lung disease,”
organisms grow slowly in culture. Up to 6 weeks of incuba- a hypersensitivity pneumonitis), cervical lymphadenitis, and
tion may be required for visible growth. MAC species are then disseminated disease.60 Disseminated disease occurs most
identified using commercial DNA probe-based assays or PCR often in HIV-infected humans, but children with congenital
CHAPTER 44  Mycobacterial Infections 429

A B
FIGURE 44-12  A, Granulomatous inflammation in the brain of a 10-year-old male neutered Siamese mix cat with disseminated Mycobacterium avium infection. H&E stain. B, Section
from the same cat stained with Ziehl-Neelsen (acid-fast) stain. There are large numbers of intracellular acid-fast bacteria.

immunodeficiencies can also be affected. Prophylactic antimyco- Inoculation of organisms into the skin through rodent bites or cat
bacterial drug treatment is recommended for HIV-infected humans fight wounds is the suspected mode of transmission.
with CD4+ T cell counts that are less than 50 cells/mm3.60 Cervi- Disease in dogs is known as canine leproid granuloma syn-
cal lymphadenitis is an ­uncommon disease of children less than drome (CLGS). It has mainly been reported from the western
3 years of age. United States and coastal regions of Australia.64-66 The organ-
Although transmission of MAC organisms from diseased ism that causes CLGS has only been partially characterized by
dogs and cats to humans should not, in theory, occur by direct the use of PCR sequencing. It has never been isolated in culture
contact, it may not be possible to distinguish infection with and does not yet have a Latin name, but is often referred to as
M. avium from infection with MTBC organisms (especially the “CLG organism.” A similar, and possibly the same, organ-
M. bovis) based on clinical presentation. Thus, in regions where ism has also been detected using PCR in cats with leprosy.63
M. bovis infections occur, animals suspected to have MAC Boxer dogs or their crosses are predisposed, and possibly also
infections should be isolated until the results of PCR assays con- German shepherd dogs.65,66 The mode of transmission to dogs
firm MAC infection. Cytologic or histopathologic evidence of is unknown. Transmission by biting insects has been hypoth-
large numbers of organisms also suggests MAC as opposed to esized because most lesions occur on the pinnae and head.
MTBC infection. Veterinary staff should take care to avoid nee-
dle-stick injuries or cutaneous inoculation when collecting spec- Clinical Features
imens (such as lymph node aspirates) from dogs and cats with
M. avium infections because of the possibility that disease might Lepromatous mycobacteria cause single or multiple nodular
occur after cutaneous inoculation. Sedation is recommended cutaneous or subcutaneous lesions, which may ulcerate. In cats,
whenever lymph node aspiration is indicated for an animal with lesions are usually on the head or forelimbs, but can also occur at
an unknown cause of lymphadenopathy. When mycobacterial other body sites. Affected cats are usually systemically well. Two
disease is suspected, care should also be taken not to aerosolize pathologic variants of feline disease have been described.62,63
tissues at necropsy (as occurs with electric saws). The first form, “tuberculoid feline leprosy,” is characterized
by multifocal to coalescing pyogranulomas with central case-
Lepromatous Mycobacteria ous necrosis, as well as increased numbers of lymphocytes and
plasma cells in the dermis and panniculus. Multi­nucleate giant
Etiology and Epidemiology cells may or may not be present, and acid-fast stains reveal
variable numbers of AFB, primarily in regions of necrosis. The
Lepromatous mycobacteria are highly fastidious or uncultur- second form, “lepromatous feline leprosy,” is characterized by
able mycobacteria that cause nodular cutaneous lesions in dogs pyogranulomatous dermatitis and panniculitis with sheets of
or cats. Feline leprosy is caused by Mycobacterium lepraem- epithelioid macrophages and neutrophils, moderate to abundant
urium and probably several other mycobacterial species. In fact, multinucleate giant cells, and large numbers of AFB with mini-
with the recent discovery of these other mycobacterial species, mal necrosis. Organisms that resemble M. lepraemurium can be
localized nodular cutaneous disease in cats caused by MAC detected in both these clinical forms, whereas the CLG organism
organisms, or organisms that resemble other nontuberculous has only been detected in cats with the tuberculoid form.63 The
mycobacteria (NTM), has fallen under the general heading of distribution of histopathologic types may vary geographically.
feline leprosy syndromes.61-63 In dogs, CLGS is characterized by the development of firm
Feline leprosy has been described in the Netherlands, the United cutaneous nodules that range in diameter from millimeters to
Kingdom, the United States, western Canada, Australia, New several centimeters. Larger lesions may ulcerate. Most lesions
Zealand, Italy, and Greece. Affected cats range in age from 1 year are found on the pinnae, but they may also be found on the head,
to 14 years and are often from rural or semirural environments. muzzle, forelimbs, and trunk (Figure 44-13). Histopathology
430 SECTION 2  Bacterial Diseases

B C
FIGURE 44-13  A, Pinna of a 5-year-old male neutered boxer dog with CLGS. A crusted lesion was present that measured 1 × 2 cm in diameter. B, Histopathology of CLGS lesions
reveals pyogranulomatous inflammation. C, A BCG immunostain (brown) identified the presence of intracellular Mycobacterium spp. organisms.

reveals coalescing accumulations of histiocytes and neutrophils, lesions, but has the potential to be followed by dehiscence and
with fewer numbers of lymphocytes and plasma cells.64,66 Mul- extension of disease. If necessary, antimicrobial drug treatment
tinucleate giant cells may be present, and there may be large for several weeks followed by surgery may be most successful.
numbers of intracellular AFB. In dogs, granulomas may resolve spontaneously within 1 to
3 months. In dogs with progressive or widespread CLGS, a com-
Diagnosis bination of clarithromycin and a rifamycin may be effective.67

Diagnosis of leproid syndromes is based on the presence of typi- Prevention and Public Health Aspects
cal skin lesions, absence of signs of dissemination or systemic
illness, suggestive cytologic and histopathologic findings, nega- Mycobacterial organisms that cause canine and feline leproma-
tive mycobacterial culture results, and positive 16S rRNA gene tous disease are not transmitted directly to humans. However,
PCR assay results where PCR assays are available. Cytologic lesions may resemble those caused by M. microti and M. bovis,
examination of fine-needle aspirates usually reveals pyogranulo- so where these diseases are endemic, precautions should be taken
matous inflammation with intracellular, negatively stained bac- until a causative organism is identified with molecular methods.
teria, which subsequently stain with acid-fast stains. Sequence
analysis of PCR products that are generated from biopsy speci-
mens is required to determine the likely species present.
Nontuberculous Bacteria Other Than
Mycobacterium avium-intracellulare
Treatment
Etiology and Epidemiology
Treatment of feline leproid lesions with a combination of clo-
fazimine and clarithromycin is often successful (see Table 44-4). NTM species include a variety of opportunistic, environmen-
Wide surgical excision could be considered for small, localized tal mycobacteria that have been classified based on their rate
CHAPTER 44  Mycobacterial Infections 431

TABLE 44-5
Cutaneous and Disseminated Disease in Dogs and Cats Caused by Slow-Growing Opportunistic Nontuberculous Mycobacteria
Mycobacterial Species ­
Species Affected Clinical Abnormalities Geographic Location
M. kansasii Dog Chronic pleural effusion with pulmonary arterial and pleuro- North Carolina, USA83
peritoneal shunt infection
M. simiae Cat Disseminated disease with generalized lymphadenopathy, lung Switzerland84
and skin involvement, and granulomatous chorioretinitis
M. genavense Cat (FIV+) Disseminated disease with involvement of lungs, liver, lymph New South Wales, Australia85
nodes, skin, kidney, spleen
Dog Disseminated disease with involvement of lymph nodes and New York, USA86
possibly liver and spleen
M. xenopi Cat Cutaneous lesion Australia87
Cat Disseminated disease with kidney, lymph node, pancreatic, Florida, USA88
and marrow involvement
Cat Disseminated disease with lymphadenopathy, gastrointestinal Wisconsin, USA89
involvement, and ascites
M. xenopi-like Cat (FIV+) Tracheal granuloma Italy90
M. ulcerans Dog Cutaneous ulcerating lesions on distal limbs or tail base Victoria, Australia91
Cat Mass on nasal bridge Victoria, Australia92
‘M. visibile’ Cat Cutaneous and disseminated disease with involvement of the Western Canada and United
(­originally eyes, skeletal muscle, and a variety of other organs except States93
‘M. visibilis’) the liver and kidneys
M. malmoense Cat Cutaneous involvement United Kingdom10
M. celatum Cat Cutaneous involvement and fever United Kingdom10
M. terrae Cat Cutaneous involvement United Kingdom94

of growth in culture (rapid, intermediate, or slow) (see Table M. goodii.72-75 No age, breed, or sex predisposition has been
44-1). They are found in soil and a variety of water sources and recognized, but most dogs are in good body condition or obese.
cause cutaneous, pulmonary, or disseminated disease in dogs Some dogs have a history of bite wounds or trauma.73 Concur-
and cats. Rapidly growing mycobacteria (RGM) grow in cul- rent hyperadrenocorticism was described in one dog.75
ture within 7 days and include several groups of organisms to M. fortuitum can cause pulmonary disease in dogs76-79 as
which M. fortuitum, M. septicum, M. abscessus, M. chelonae, well as bacteremia.72 Lipoid mycobacterial pneumonia rarely
M. smegmatis, M. thermoresistibile, M. goodii, and M. muco- occurs in cats and may arise in association with lactulose or par-
genicum belong. Slowly growing mycobacteria require more affin treatment for furballs.80 Pneumonia was also described in
than 7 days (and sometimes many months) before significant a cat infected with M. thermoresistibile.81 Disseminated RGM
growth occurs in culture. Other than MAC organisms, exam- infections are very rare. Disseminated disease that resembled
ples include M. kansasii, M. terrae, M. xenopi, M. ulcerans, MAC infection was described in a basset hound infected with
M. malmoense, M. simiae, and M. genavense. M. smegmatis.82

Rapidly Growing Mycobacteria Slowly Growing Mycobacteria


RGM are responsible for the vast majority of cutaneous myco- A variety of slowly growing mycobacteria cause cutaneous or dis-
bacterial disease in cats in the United States and Australia, but seminated disease in dogs and cats that resembles disease caused
infections have also been described in the United Kingdom, by MAC organisms (Table 44-5).83-94 These include the known
Canada, western Europe, and New Zealand. The most com- human pathogens M. kansasii, M. simiae, M. genavense, M.
monly isolated species in both the United States and Australia xenopi, M. celatum, M. malmoense, M. terrae, and M. ulcerans.
are M. fortuitum and M. smegmatis, but infections with other Some, but not all affected cats have been infected with FIV, and
RGM also occur (see Table 44-1).68-71 These organisms have a one cat had CD4+ lymphopenia.88 M. ulcerans causes ulcerative
tropism for adipose tissue, and when inoculated into the skin skin lesions in humans, cats, or dogs in certain parts of Australia.
by means of cat fight injuries (bites or scratches) or other pen-
etrating wounds (and rarely surgical wounds),72 mycobacterial Clinical Features
panniculitis develops. Most affected cats are female spayed cats.
Underlying immunosuppressive disease is usually not present. Rapidly Growing Mycobacteria
Cutaneous disease occurs less often in dogs and results from In cats, mycobacterial panniculitis occurs most often in the
infection with M. fortuitum, M. smegmatis, M. abscessus, or inguinal fat pad, but lesions may also be found elsewhere
432 SECTION 2  Bacterial Diseases

on the lateral abdomen, perineum, dorsum, and pelvic limbs contained mostly neutrophils.76 Heavily vacuolated macrophages
(Figure 44-14). The subcutis becomes thickened and adheres to and lipid vacuoles may be present in aspirates from animals with
the dermis, after which nodular and draining skin lesions develop. panniculitis. Organisms may or may not be visible with routine
The majority of affected cats are otherwise well; the development stains or acid-fast stains. Histopathology shows pyogranulo-
of systemic illness is rare. In dogs, nodular lesions or nonhealing matous inflammation without tubercle formation or necrosis.
wounds with draining tracts often occur in the inguinal region, Large lipid vacuoles that contain acid-fast bacilli may be present.
cervical region, and/or other proximal body locations. Although molecular assays are often used for speciation, direct
Pneumonia due to RGM generally results in signs of fever, use of PCR assays on clinical specimens is often not required for
lethargy, decreased appetite, cough, and tachypnea, with diagnosis because RGM grow readily on routine culture media
increased lung sounds on thoracic auscultation. Thoracic radio- (blood agar). Culture also allows subsequent antimicrobial drug
graphs frequently reveal lung lobe consolidation and alveolar susceptibility testing to be performed at specialized facilities.
patterns. In the cat with M. thermoresistibile pneumonia, exten- Cytology and histopathology findings for slow-growing
sive pulmonary mineralization developed.81 The dog with dis- NTM are similar to those for MAC infections. Diagnosis of
seminated M. smegmatis infection had granulomatous hepatitis slow-growing mycobacterial infection requires specialized
and lymphadenitis. mycobacterial culture or the use of PCR assays. M. xenopi
requires culture at higher temperatures (42°C to 44°C), whereas
Slowly Growing Mycobacteria Other Than MAC other mycobacterial species require lower temperatures (28°C
Clinical features of slowly growing mycobacterial infections or 30°C). Organisms such as M. genavense and M. ulcerans can
other than MAC infections have varied from one case to another be difficult to grow on solid media, and so molecular methods
and are summarized in Table 44-5. Animals with disseminated may be required to identify these species.
disease can have clinical abnormalities that are indistinguish-
able from those caused by M. avium. Treatment and Prognosis
Diagnosis Treatment options for RGM infections include doxycycline, flu-
oroquinolones, clarithromycin, aminoglycosides, sulfonamides,
Diagnosis of RGM infection is usually based on the presence of or carbapenems (see Table 44-4 and Chapter 8). Many cutane-
consistent clinical abnormalities, and the results of cytology, his- ous and pulmonary infections respond well to treatment with
topathology, and mycobacterial culture (see Table 44-3). Cyto- doxycycline, aminoglycosides, and/or a fluoroquinolone.74-78
logic examination of fine needle aspirates or lavage specimens Multidrug-resistant isolates are more difficult to treat; moxi-
generally reveals pyogranulomatous inflammation. However, floxacin, pradofloxacin, or linezolid could be considered when
airway lavage fluid from a dog with M. fortuitum pneumonia other treatment options are not possible.95 As for lepromatous

A C
FIGURE 44-14  Cutaneous-subcutaneous infections with rapidly-growing mycobacteria. A and B, Thirteen-year-old male neutered Siamese cat with mycobacterial panniculitis. There
are multiple nodular lesions in the inguinal region (A) and lateral flank and dorsal lumbar regions (B). In this cat, lesions progressed slowly over 8 years despite treatment with antimicrobial
drugs. C, Dorsum of an 8-year old dachshund mix with a multidrug resistant Mycobacterium abscessus infection. Multiple draining wounds and extensive scarring are present.
CHAPTER 44  Mycobacterial Infections 433

disease, surgical excision of cutaneous lesions is often followed prognosis for disseminated infections in dogs and cats appears
by dehiscence and progression if resection is not complete. to be guarded to poor.
However, radical surgical excision by an experienced surgeon
could be considered after antimicrobial drug treatment if lesions Public Health Aspects
reduce in size but fail to resolve. Intraoperative treatment of
affected cats with an aminoglycoside has been suggested.96 NTM are opportunistic, environmental mycobacteria that are
Treatment should continue for 2 months beyond complete reso- not usually transmitted from animals to humans. Disease in
lution of the lesions and is generally required for at least 4 to humans resembles that in dogs and cats. Localized cutane-
12 months. ous and soft tissue infections with NTM have occurred after
M. ulcerans infections generally respond to monotherapy traumatic wounds and lacerations, including after dog and cat
with clarithromycin or a fluoroquinolone.91,92 For animals bites.98-100 Infections with RGM can occur after surgical proce-
with slowly growing mycobacterial infections, options similar dures such as liposuction, LASIK surgery, and acupuncture.97
to those used to treat MAC infections are indicated—that is, Catheter-related infections have also been described. Dissemi-
combinations of clarithromycin, a rifamycin, and a fluoroqui- nated disease caused by NTM almost always occurs in humans
nolone. In human patients, M. kansasii infections are treated with HIV infection or other immunosuppressive illness or drug
with rifampin and ethambutol, with or without isoniazid.97 The therapy.

CASE EXAMPLE Respiratory: Referred upper airway noises were auscultated.


Gastrointestinal/Genitourinary: The dog’s abdomen
was nonpainful. The rectal mucosa was proliferative
Signalment: “Sarah”, a 5-year-old female spayed husky dog and hyperemic and protruded from the anus. A rectal
from Santa Cruz in coastal northern California examination revealed a nodular diffusely thickened mucosa
History: Sarah was evaluated for a 1-month history of with a granular texture and prominent sublumbar lymph
protrusion of the anus, swelling of the neck, progressive nodes. There was evidence of melena and frank blood on the
lethargy, and inappetence. She had been diagnosed with glove. No vulvar discharge was noted.
zinc-responsive dermatitis 3 years prior, and symmetrical Neurologic: Palpebral, menace, and papillary light responses
lupoid onychodystrophy 6 months prior. There were two were intact bilaterally. No neurologic deficits were appa­rent. A
other dogs and three cats in the household, none of which full neurologic examination was not performed.
were unwell. The dogs lived in a suburban household and Lymph Nodes: Generalized peripheral lymphadenopathy was
occasionally traveled to southern California, and were present. The mandibular nodes were 5-6 cm in diameter,
normally fed a commercial dry maintenance dog food. lobular and firm. The superficial cervical nodes were 3 cm
There was no known history of tick or toxin exposure or in diameter, and the inguinal nodes were enlarged at 2 cm,
interactions with other wildlife or domestic farm animal with the left larger than the right. The popliteal nodes were
species. The owner was a white non-Hispanic male who enlarged at 2-3 cm bilaterally.
had been sick for some months. His doctor believed he had Laboratory Findings:
acquired the illness from Sarah. CBC:
Current Medications: Zinc (2.5 mg/kg q12h PO), tetracycline HCT 22% (40%-55%)
(25 mg/kg q12h PO), niacinamide (25 mg/kg q12h PO), MCV 52 fL (65-75 fL)
fish oil capsules (1 capsule PO q12h), and vitamin E (20 MCHC 30 g/dL (33-36 g/dL)
IU/kg PO q12h) for the zinc-responsive dermatitis and Reticulocyte count 25,200 cells/µL
onychodystrophy. Fipronil with S-methoprene was used for WBC 40,510 cells/µL (6000-13,000 cells/µL)
flea and tick control on a monthly basis. Neutrophils 29,977 cells/µL (3000-10,500 cells/µL)
Physical Examination: Band neutrophils 2431 cells/µL
Body Weight: 20 kg Lymphocytes 2026 cells/µL (1000-4000 cells/µL)
General: Quiet to lethargic, hydrated, responsive, preferred to Monocytes 2431 cells/µL (150-1200 cells/µL)
lie down but was ambulatory. T=102.9°F (39.4°C), HR = 140 Eosinophils 3646 cells/µL (0-1500 cells/µL)
beats/min, RR = 70 breaths/min Platelets 387,000 platelets/µL (150,000-400,000 platelets/µL).
Integument: No abnormalities were detected. Band and mature neutrophils were slightly toxic.
Eyes, Ears, Nose, and Throat: A small amount of purulent Serum Chemistry Profile:
ocular discharge was present from the left eye. A fundic Sodium 132 mmol/L (145-154 mmol/L)
examination was unremarkable. Moderate dental calculus. Potassium 3.5 mmol/L (3.6-5.3 mmol/L)
Musculoskeletal: Body condition score was 4/9, with Chloride 103 mmol/L (108-118 mmol/L)
symmetrical muscling. Moderate ventral cervical and Bicarbonate 17 mmol/L (16-26 mmol/L)
submandibular swelling was present. Phosphorus 4.1 mg/dL (3.0-6.2 mg/dL)
Cardiovascular: No murmurs or arrhythmias, femoral pulses Calcium 8.6 mg/dL (9.7-11.5 mg/dl)
adequate. Mucous membranes were pale pink, with a CRT of 2 s. BUN 6 mg/dL (5-21 mg/dL)

Continued
434 SECTION 2  Bacterial Diseases

Creatinine 0.6 mg/dL (0.3-1.2 mg/dL) Real-time PCR assay for M. avium complex organisms (lymph
Glucose 93 mg/dL (64-123 mg/dL) node aspirate): Positive
Total protein 4.3 g/dL (5.4-7.6 g/dL) Mycobacterial culture and susceptibility testing (National Jew-
Albumin 2.1 g/dL (3.0-4.4 g/dL) ish Laboratories, Denver, CO): Positive for M. avium after in-
Globulin 2.2 g/dL (1.8-3.9 g/dL) cubation for 26 days. The organism was identified by HPLC.
ALT 43 U/L (19-67 U/L) The organism was tentatively designated as resistant to
AST 51 U/L (19-42 U/L) ciprofloxacin, rifabutin, and rifampin; of intermediate sus-
ALP 115 U/L (21-170 U/L) ceptibility to clofazimine, kanamycin, amikacin, ethambutol,
Creatine kinase 202 U/L (51-399 U/L) streptomycin, and moxifloxacin; and susceptible to clar-
GGT < 3 U/L (0-6 U/L) ithromycin. The organism was susceptible to ethambutol
Cholesterol 146 mg/dL (135-361 mg/dL) and of intermediate susceptibility to rifampin when the two
Total bilirubin 0 mg/dL (0-0.2 mg/dL) drugs were used in combination.
Magnesium 2.0 mg/dL (1.5-2.6 mg/dL). Diagnosis: Disseminated M. avium infection.
Urinalysis: SGr 1.005; pH 7.5; no protein, bilirubin, hemoprotein, Treatment: Before the availability of culture results, the
or glucose; rare WBC/HPF, 0 RBC/HPF, few lipid droplets. dog was treated with a combination of enrofloxacin,
Imaging Findings: clarithromycin, and ethambutol. The tetracycline and
Thoracic Radiographs: No abnormalities were detected. niacinamide combination was discontinued because of the
Abdominal Ultrasound: All abdominal lymph nodes potential for underlying immunosuppression. The lymph
were enlarged and irregular. There were focal regions of nodes ruptured after the aspirates were collected and
thickening and irregularity within the small bowel. The liver drained purulent exudate. Unfortunately, this was followed
and spleen were unremarkable. shortly afterward by the development of fever, lethargy, and
Aspiration Cytology (Mandibular Lymph Node): dysphagia secondary to severe lymphadenomegaly, and the
Multiple, variably sized aggregates of macrophages were owners elected euthanasia.
scattered along with numerous karyolytic and degenerated Comments: This dog was suspected to have mycobacterial
neutrophils and a residual lymphoid population in a light pink infection caused by an organism other than a MTBC organism
background that contained low numbers of erythrocytes from the outset, based on the dog’s background, clinical
and lysed cells. The macrophages were large and distended presentation, and the presence of large numbers of AFB on
with intracellular, negatively staining thick, relatively short, cytology. M. tuberculosis most often causes pulmonary disease
rod-shaped bacteria. Intracellular organisms were noted in dogs. M. bovis infections have been described in the Hispanic
within neutrophils and large multinucleated giant cells, as population in southern California and rarely in cattle from
well as being located extracellularly. The bacilli were acid California,101 so M. bovis infection was possible but seemed
fast. Interpretation: Pyogranulomatous inflammation due to highly unlikely based on exposure history and the epidemiology
infection with Mycobacterium spp. of this disease in dogs. The use of PCR assays allowed rapid
Microbiologic Testing: Real-time PCR assay for Mycobacter­ium confirmation that the organism belonged to M. avium.
genus organisms (lymph node aspirate): Positive

SUGGESTED READINGS 4. Hackendahl NC, Mawby DI, Bemis DA, et al. Putative transmis-
sion of Mycobacterium tuberculosis infection from a human to a
Baral RM, Metcalfe SS, Krockenberger MB, et al. Disseminated Myco- dog. J Am Vet Med Assoc. 2004;225:1573-1577:1548.
bacterium avium infection in young cats: overrepresentation of Abys- 5. Liu S, Weitzman I, Johnson GG. Canine tuberculosis. J Am Vet
sinian cats. J Feline Med Surg. 2006;8:23-44. Med Assoc. 1980;177:164-167.
Bennett S. Photosensitisation induced by clofazimine in a cat. Aust Vet J. 6. Foster ES, Scavelli TD, Greenlee PG, et  al. Cutaneous lesion
2007;85:375-380. caused by Mycobacterium tuberculosis in a dog. J Am Vet Med
Davies JL, Sibley JA, Myers S, et al. Histological and genotypical character- Assoc. 1986;188:1188-1190.
ization of feline cutaneous mycobacteriosis: a retrospective study of for- 7. Sykes JE, Cannon AB, Norris AJ, et  al. Mycobacterium
malin-fixed paraffin-embedded tissues. Vet Dermatol. 2006;17:155-162. tuberculosis complex infection in a dog. J Vet Intern Med.
Posthaus H, Bodmer T, Alves L, et al. Accidental infection of veterinary 2007;21:1108-1112.
personnel with Mycobacterium tuberculosis at necropsy: a case study. 8. Parsons SD, Gous TA, Warren RM, et al. Pulmonary Mycobacte-
Vet Microbiol. 2011;149:374-380. rium tuberculosis (Beijing strain) infection in a stray dog. J S Afr
Shrikrishna D, de la Rua-Domenech R, Smith NH, et  al. Human and Vet Assoc. 2008;79:95-98.
canine pulmonary Mycobacterium bovis infection in the same house- 9. Erwin PC, Bemis DA, McCombs SB, et al. Mycobacterium tuber-
hold: re-emergence of an old zoonotic threat? Thorax. 2009;64:89-91. culosis transmission from human to canine. Emerg Infect Dis.
2004;10:2258–2260.
REFERENCES 10. Gunn-Moore DA, McFarland SE, Brewer JI, et al. Mycobacterial
1. Sakula A. Robert Koch: centenary of the discovery of the tubercle disease in cats in Great Britain: I. Culture results, geographical
bacillus, 1882. Thorax. 1982;37:246-251. distribution and clinical presentation of 339 cases. J Feline Med
2. Posthaus H, Bodmer T, Alves L, et al. Accidental infection of vet- Surg. 2011;13:934-944.
erinary personnel with Mycobacterium tuberculosis at necropsy: 11. de Lisle GW, Collins DM, Loveday AS, et al. A report of tuber-
a case study. Vet Microbiol. 2011;149:374-380. culosis in cats in New Zealand, and the examination of strains of
3. Turinelli V, Ledieu D, Guilbaud L, et  al. Mycobacterium Mycobacterium bovis by DNA restriction endonuclease analysis.
­tuberculosis infection in a dog from Africa. Vet Clin Pathol. N Z Vet J. 1990;38:10-13.
2004;33:177-181.
CHAPTER 44  Mycobacterial Infections 435

12. Zumarraga MJ, Vivot MM, Marticorena D, et al. Mycobacterium 35. Campora L, Corazza M, Zullino C, et al. Mycobacterium avium
bovis in Argentina: isolates from cats typified by spoligotyping. subspecies hominissuis disseminated infection in a Basset Hound
Rev Argent Microbiol. 2009;41:215-217. dog. J Vet Diagn Invest. 2011;23:1083-1087.
13. Kaneene JB, Bruning-Fann CS, Dunn J, et al. Epidemiologic inves- 36. de Groot PH, van Ingen J, de Zwaan R, et al. Disseminated Myco-
tigation of Mycobacterium bovis in a population of cats. Am J Vet bacterium avium subsp. avium infection in a cat, the Netherlands.
Res. 2002;63:1507-1511. Vet Microbiol. 2010;144:527-529.
14. Corner LA, Murphy D, Gormley E. Mycobacterium bovis infec- 37. Glanemann B, Schonenbrucher H, Bridger N, et  al. Detection
tion in the Eurasian badger (Meles meles): the disease, pathogen- of Mycobacterium avium subspecies paratuberculosis–specific
esis, epidemiology and control. J Comp Pathol. 2011;144:1-24. DNA by PCR in intestinal biopsies of dogs. J Vet Intern Med.
15. Wilkins MJ, Bartlett PC, Berry DE, et al. Absence of Mycobacte- 2008;22:1090-1094.
rium bovis infection in dogs and cats residing on infected cattle 38. Whiley H, Keegan A, Giglio S, et al. Mycobacterium avium com-
farms: Michigan, 2002. Epidemiol Infect. 2008;136:1617-1623. plex—the role of potable water in disease transmission. J Appl
16. van der Burgt GM, Crawshaw T, Foster AP, et al. Mycobacterium Microbiol. 2012;113:223-232.
bovis infection in dogs. Vet Rec. 2009;165:634. 39. Mijs W, de Haas P, Rossau R, et al. Molecular evidence to sup-
17. Ellis MD, Davies S, McCandlish IA, et al. Mycobacterium bovis port a proposal to reserve the designation Mycobacterium avium
infection in a dog. Vet Rec. 2006;159:46-48. subsp. avium for bird-type isolates and “M. avium subsp. hom-
18. Shrikrishna D, de la Rua-Domenech R, Smith NH, et al. Human inissuis” for the human/porcine type of M. avium. Int J Syst Evol
and canine pulmonary Mycobacterium bovis infection in the Microbiol. 2002;52:1505-1518.
same household: re-emergence of an old zoonotic threat? Thorax. 40. Jordan HL, Cohn LA, Armstrong PJ. Disseminated Mycobacte-
2009;64:89-91. rium avium complex infection in three Siamese cats. J Am Vet
19. Smith NH, Crawshaw T, Parry J, et al. Mycobacterium microti: Med Assoc. 1994;204:90-93.
More diverse than previously thought. J Clin Microbiol. 41. Baral RM, Metcalfe SS, Krockenberger MB, et  al. Disseminated
2009;47:2551-2559. Mycobacterium avium infection in young cats: overrepresentation
20. Rufenacht S, Bogli-Stuber K, Bodmer T, et  al. Mycobacterium of Abyssinian cats. J Feline Med Surg. 2006;8:23-44.
microti infection in the cat: a case report, literature review and 42. Griffin A, Newton AL, Aronson LR, et al. Disseminated Mycobac-
recent clinical experience. J Feline Med Surg. 2011;13:195-204. terium avium complex infection following renal transplantation in
21. Deforges L, Boulouis HJ, Thibaud JL, et  al. First isolation of a cat. J Am Vet Med Assoc. 2003;222:1097-1101:1077-1098.
Mycobacterium microti (Llama-type) from a dog. Vet Microbiol. 43. Kadar E, Sykes JE, Kass PH, et al. Evaluation of the prevalence
2004;103:249-253. of infections in cats after renal transplantation: 169 cases (1987-
22. Bauer NB, O’Neill E, Sheahan BJ, et al. Calcospherite-like bodies 2003). J Am Vet Med Assoc. 2005;227:948-953.
and caseous necrosis in tracheal mucus from a dog with tubercu- 44. Malik R, Gabor L, Martin P, et  al. Subcutaneous granuloma
losis. Vet Clin Pathol. 2004;33:168-172. caused by Mycobacterium avium complex infection in a cat. Aust
23. Gunn-Moore DA, Jenkins PA, Lucke VM. Feline tuberculosis: a Vet J. 1998;76:604-607.
literature review and discussion of 19 cases caused by an unusual 45. Graham KJ, Brain PH, Spielman D, et  al. Concurrent infection
mycobacterial variant. Vet Rec. 1996;138:53-58. with Cryptococcus neoformans/gattii species complex and Myco-
24. de Bolla GJ. Tuberculosis in a cat. Vet Rec. 1994;134:336. bacterium avium affecting the subcutis and bone of a pelvic limb
25. Blunden AS, Smith KC. A pathological study of a mycobacterial in a cat. J Feline Med Surg. 2011;13:776-780.
infection in a cat caused by a variant with cultural characteris- 46. Riviere D, Pingret JL, Etievant M, et al. Disseminated Mycobac-
tics between Mycobacterium tuberculosis and M. bovis. Vet Rec. terium avium subspecies infection in a cat. J Feline Med Surg.
1996;138:87-88. 2011;13:125-128.
26. Bennett AD, Lalor S, Schwarz T, et al. Radiographic findings in cats 47. Barry M, Taylor J, Woods JP. Disseminated Mycobacterium
with mycobacterial infections. J Feline Med Surg. 2011;13:718-724. avium infection in a cat. Can Vet J. 2002;43:369-371.
27. Greene CE, Gunn-Moore DA. Infections caused by slow-growing 48. Sieber-Ruckstuhl NS, Sessions JK, Sanchez S, et  al. Long-term
mycobacteria. In: Greene CE, ed. Infectious Diseases of the Dog cure of disseminated Mycobacterium avium infection in a cat. Vet
and Cat. 4th ed. St. Louis, MO: Elsevier Saunders; 2012:495-510. Rec. 2007;160:131-132.
28. Rhodes SG, Gruffydd-Jones T, Gunn-Moore D, et al. Adaptation 49. Morita Y, Kimur H, Kozawa K, et  al. Cutaneous infection in a
of IFN-gamma ELISA and ELISPOT tests for feline tuberculosis. cat caused by Mycobacterium avium complex serovar 6. Vet Rec.
Vet Immunol Immunopathol. 2008;124:379-384. 2003;152:120.
29. Saleeb PG, Drake SK, Murray PR, et al. Identification of myco- 50. Latimer KS, Jameson PH, Crowell WA, et al. Disseminated Myco-
bacteria in solid-culture media by matrix-assisted laser desorp- bacterium avium complex infection in a cat: presumptive diag-
tion ionization-time of flight mass spectrometry. J Clin Microbiol. nosis by blood smear examination. Vet Clin Pathol. 1997;26:
2011;49:1790-1794. 85-89.
30. Gunn-Moore DA, McFarland SE, Schock A, et al. Mycobacterial 51. Miller MA, Greene CE, Brix AE. Disseminated Mycobacterium
disease in a population of 339 cats in Great Britain: II. Histopa- avium-intracellulare complex infection in a miniature schnauzer.
thology of 225 cases, and treatment and outcome of 184 cases. J Am Anim Hosp Assoc. 1995;31:213-216.
J Feline Med Surg. 2011;13:945-952. 52. Gow AG, Gow DJ. Disseminated Mycobacterium avium complex
31. Xavier Emmanuel F, Seagar AL, Doig C, et al. Human and animal infection in a dog. Vet Rec. 2008;162:594-595.
infections with Mycobacterium microti. Scotland. Emerg Infect 53. Kim DY, Cho DY, Newton JC, et  al. Granulomatous myeli-
Dis. 2007;13:1924-1927. tis due to Mycobacterium avium in a dog. Vet Pathol.
32. World Health Organization. World Health Organization Report 1994;31:491-493.
on the Global Tuberculosis Epidemic. Geneva; 2012. Available 54. Horn B, Forshaw D, Cousins D, et al. Disseminated Mycobacte-
from http://www.who.int/tb/publications/global_report/en/. Last rium avium infection in a dog with chronic diarrhoea. Aust Vet J.
accessed February 2, 2013. 2000;78:320-325.
33. Trends in tuberculosis-United States, 2011. MMWR Morb Mor- 55. Bauer N, Burkhardt S, Kirsch A, et al. Lymphadenopathy and diar-
tal Wkly Rep 2012;61:181–185. rhea in a Miniature Schnauzer. Vet Clin Pathol. 2002;31:61-64.
34. Haist V, Seehusen F, Moser I, et al. Mycobacterium avium subsp. 56. O’Toole D, Tharp S, Thomsen BV, et  al. Fatal mycobacteriosis
hominissuis infection in 2 pet dogs. Germany. Emerg Infect Dis. with hepatosplenomegaly in a young dog due to Mycobacterium
2008;14:988-990. avium. J Vet Diagn Invest. 2005;17:200-204.
436 SECTION 2  Bacterial Diseases

57. Eggers JS, Parker GA, Braaf HA, et  al. Disseminated Mycobac- 79. Jang SS, Eckhaus MA, Saunders G. Pulmonary Mycobacterium
terium avium infection in three miniature schnauzer litter mates. fortuitum infection in a dog. J Am Vet Med Assoc. 1984;184:
J Vet Diagn Invest. 1997;9:424-427. 96-98.
58. Naughton JF, Mealey KL, Wardrop KJ, et al. Systemic Mycobacteri­um 80. Couto SS, Artacho CA. Mycobacterium fortuitum pneumonia in a
avium infection in a dog diagnosed by polymerase chain reac­tion cat and the role of lipid in the pathogenesis of atypical mycobacte-
analysis of buffy coat. J Am Anim Hosp Assoc. 2005;41:128-132. rial infections. Vet Pathol. 2007;44:543-546.
59. Radomski N, Thibault VC, Karoui C, et al. Determination of geno- 81. Foster SF, Martin P, Davis W, et al. Chronic pneumonia caused
typic diversity of Mycobacterium avium subspecies from human and by Mycobacterium thermoresistibile in a cat. J Small Anim Pract.
animal origins by mycobacterial interspersed repetitive-unit-variable- 1999;40:433-438.
number tandem-repeat and IS1311 restriction fragment length poly- 82. Grooters AM, Couto CG, Andrews JM, et  al. Systemic Myco-
morphism typing methods. J Clin Microbiol. 2010;48:1026-1034. bacterium smegmatis infection in a dog. J Am Vet Med Assoc.
60. Gordin FM, Horsburgh CR. Mycobacterium avium complex. 1995;206:200-202.
In: Mandell GL, Bennett JE, Dolin R, eds. Principles and Prac- 83. Pressler BM, Hardie EM, Pitulle C, et  al. Isolation and iden-
tice of Infectious Diseases. 7th ed. Philadelphia, PA: Elsevier; tification of Mycobacterium kansasii from pleural fluid of a
2010:3177-3189. dog with persistent pleural effusion. J Am Vet Med Assoc.
61. Hughes MS, James G, Taylor MJ, et al. PCR studies of feline lep- 2002;220:1313-1334;1336-1340.
rosy cases. J Feline Med Surg. 2004;6:235-243. 84. Dietrich U, Arnold P, Guscetti F, et  al. Ocular manifestation of
62. Malik R, Hughes MS, James G, et al. Feline leprosy: two different disseminated Mycobacterium simiae infection in a cat. J Small
clinical syndromes. J Feline Med Surg. 2002;4:43-59. Anim Pract. 2003;44:121-125.
63. Davies JL, Sibley JA, Myers S, et al. Histological and genotypical 85. Hughes MS, Ball NW, Love DN, et al. Disseminated Mycobacte-
characterization of feline cutaneous mycobacteriosis: a retrospec- rium genavense infection in a FIV-positive cat. J Feline Med Surg.
tive study of formalin-fixed paraffin-embedded tissues. Vet Der- 1999;1:23-29.
matol. 2006;17:155-162. 86. Kiehn TE, Hoefer H, Bottger EC, et al. Mycobacterium genavense
64. Charles J, Martin P, Wigney DI, et  al. Cytology and histopa- infections in pet animals. J Clin Microbiol. 1996;34:1840-1842.
thology of canine leproid granuloma syndrome. Aust Vet J. 87. Tomasovic AA, Rac R, Purcell DA. Mycobacterium xenopi in a
1999;77:799-803. skin lesion of a cat. Aust Vet J. 1976;52:103.
65. Malik R, Love DN, Wigney DI, et  al. Mycobacterial nodular 88. Meeks C, Levy JK, Crawford PC, et  al. Chronic disseminated
granulomas affecting the subcutis and skin of dogs (canine leproid Mycobacterium xenopi infection in a cat with idiopathic CD4+ T lym-
granuloma syndrome). Aust Vet J. 1998;76:403-407:398. phocytopenia. J Vet Intern Med. 2008;22:1043-1047.
66. Foley JE, Borjesson D, Gross TL, et  al. Clinical, microscopic, 89. MacWilliams PS, Whitley N, Moore F. Lymphadenitis and peri-
and molecular aspects of canine leproid granuloma in the United tonitis caused by Mycobacterium xenopi in a cat. Vet Clin Pathol.
States. Vet Pathol. 2002;39:234-239. 1998;27:50-53.
67. Malik R, Martin P, Wigney D, et al. Treatment of canine leproid 90. De Lorenzi D, Solano-Gallego L. Tracheal granuloma because of
granuloma syndrome: preliminary findings in seven dogs. Aust infection with a novel mycobacterial species in an old FIV-positive
Vet J. 2001;79:30-36. cat. J Small Anim Pract. 2009;50:143-146.
68. Malik R, Wigney DI, Dawson D, et  al. Infection of the subcu- 91. O’Brien CR, McMillan E, Harris O, et al. Localised Mycobacte-
tis and skin of cats with rapidly growing mycobacteria: a review rium ulcerans infection in four dogs. Aust Vet J. 2011;89:506-510.
of microbiological and clinical findings. J Feline Med Surg. 92. Elsner L, Wayne J, O’Brien CR, et  al. Localised Mycobacte-
2000;2:35-48. rium ulcerans infection in a cat in Australia. J Feline Med Surg.
69. Horne KS, Kunkle GA. Clinical outcome of cutaneous rapidly 2008;10:407-412.
growing mycobacterial infections in cats in the south-eastern 93. Appleyard GD, Clark EG. Histologic and genotypic characteriza-
United States: a review of 10 cases (1996-2006). J Feline Med tion of a novel Mycobacterium species found in three cats. J Clin
Surg. 2009;11:627-632. Microbiol. 2002;40:2425-2430.
70. Jang SS, Hirsh DC. Rapidly growing members of the genus 94. Henderson SM, Baker J, Williams R, et al. Opportunistic myco-
Mycobacterium affecting dogs and cats. J Am Anim Hosp Assoc. bacterial granuloma in a cat associated with a member of the
2002;38:217-220. Mycobacterium terrae complex. J Feline Med Surg. 2003;5:37-41.
71. Beccati M, Peano A, Gallo MG. Pyogranulomatous panniculi- 95. Govendir M, Hansen T, Kimble B, et al. Susceptibility of rapidly
tis caused by Mycobacterium alvei in a cat. J Small Anim Pract. growing mycobacteria isolated from cats and dogs, to ciprofloxacin,
2007;48:664. enrofloxacin and moxifloxacin. Vet Microbiol. 2011;147:113-118.
72. Sykes JE. Unpublished observations, 2012. 96. O’Brien CR, Fyfe JA, Malik R. Infections caused by rapidly-­
73. Malik R, Shaw SE, Griffin C, et al. Infections of the subcutis and growing mycobacteria. In: Greene CE, ed. Infectious Diseases of
skin of dogs caused by rapidly growing mycobacteria. J Small the Dog and Cat. 4th ed. St. Louis, MO: Elsevier Saunders; 2012:
Anim Pract. 2004;45:485-494. 515-521.
74. Bryden SL, Burrows AK, O’Hara AJ. Mycobacterium goodii infec- 97. Brown-Elliott BA, Wallace RJ. Infections due to non-tuberculous
tion in a dog with concurrent hyperadrenocorticism. Vet Derma- mycobacteria other than Mycobacterium avium-intracellulare. In:
tol. 2004;15:331-338. Mandell GL, Bennett JE, Dolin R, eds. Principles and Practice of
75. Fox LE, Kunkle GA, Homer BL, et al. Disseminated subcutane- Infectious Diseases. Philadelphia, PA: Elsevier; 2010:3191-3198.
ous Mycobacterium fortuitum infection in a dog. J Am Vet Med 98. Southern Jr PM. Tenosynovitis caused by Mycobacterium kansasii
Assoc. 1995;206:53-55. associated with a dog bite. Am J Med Sci. 2004;327:258-261.
76. Irwin PJ, Whithear K, Lavelle RB, et al. Acute bronchopneumonia 99. Ariel I, Haas H, Weinberg H, et  al. Mycobacterium fortuitum
associated with Mycobacterium fortuitum infection in a dog. Aust granulomatous synovitis caused by a dog bite. J Hand Surg Am.
Vet J. 2000;78:254-257. 1983;8:342-343.
77. Wylie KB, Lewis DD, Pechman RD, et al. Hypertrophic osteopa- 100. Ngan N, Morris A, de Chalain T. Mycobacterium fortuitum
thy associated with Mycobacterium fortuitum pneumonia in a infection caused by a cat bite. N Z Med J. 2005;118:U1354.
dog. J Am Vet Med Assoc. 1993;202:1986-1988. 101. Rodwell TC, Kapasi AJ, Moore M, et al. Tracing the origins of
78. Turnwald GH, Pechman RD, Turk JR, et  al. Survival of a dog Mycobacterium bovis tuberculosis in humans in the USA to cattle
with pneumonia caused by Mycobacterium fortuitum. J Am Vet in Mexico using spoligotyping. Int J Infect Dis. 2010;14(suppl 3):
Med Assoc. 1988;192:64-66. e129-e135.
CHAPTER 45

Salmonellosis
Jane E. Sykes and Stanley L. Marks

multiple serotypes.3 Serotypes of S. bongori are generally asso-


Overview of Salmonellosis ciated with cold-blooded animals and rarely have been isolated
First Described: Washington, DC, United States, 1885 (Theo- from warm-blooded animals, including a dog with diarrhea.4
bald Smith and Daniel Salmon)1 There are six subspecies of S. enterica (Table 45-1) and approx-
Cause: Salmonella spp. (gram-negative rods that belong to imately 2500 different serotypes. These are identified on the
the Enterobacteriaceae) basis of agglutination reactions of their O (somatic) and H
(flagellar) antigens (see Chapter 36). Subspecies I serotypes,
Affected Hosts: A large variety of warm-blooded animals
which are responsible for almost all disease in humans and
and humans; cold-blooded animals may be subclinically
warm-blooded animals worldwide, are assigned names such
infected
as S. ser. Typhimurium. Unless named before 1966, serotypes
Geographic Distribution: Worldwide that belong to other subspecies are designated using antigenic
Primary Mode of Transmission: Fecal-oral formulas. The formulas describe the subspecies, O antigens: H
antigens (phase 1): and if present, H antigens (phase 2) (e.g., S.
Major Clinical Signs: Fever, lethargy, anorexia, diarrhea, vom-
IV 45:g,z51:—).
iting, and less commonly reproductive failure, neurologic
Certain serotypes of Salmonella are host restricted, whereas
and/or respiratory signs
others infect a broad range of host species. Salmonella serotype
Differential Diagnoses: Differential diagnoses for suspected Typhi, for example, is adapted to humans, causing typhoid
Salmonella enterocolitis include canine and feline parvo- fever, and does not infect animals. Other serotypes are adapted
virus infection, canine distemper virus infection, campylo- to certain host species, with rare transmission to other hosts.
bacteriosis, clostridial diarrhea, salmon poisoning disease, Salmonella serotype Typhimurium is the most commonly iso-
giardiasis, tritrichomoniasis (cats), cryptosporidiosis, whip- lated serotype from diseased humans and animals.
worms, leptospirosis, dietary indiscretion, gastrointestinal Infection of dogs and cats with Salmonella has been associ-
foreign body, pancreatitis, inflammatory bowel disease, ated with the feeding of raw meat diets,5-9 although commercial
lymphoma, hypoadrenocorticism, hyperthyroidism, toxins dry and raw dog food and pig ear pet treats have also become
(including drugs) contaminated with the organism.5,10-13 Occasionally, co-infec-
Human Health Significance: Important zoonosis. Dogs and tions with multiple Salmonella serotypes occur.14 Outbreaks of
cats may be a source of human infection with some Sal- S. ser. Typhimurium infection in cats have been associated with
monella serotypes, some of which may be resistant to seasonal bird migrations (“songbird fever”).
multiple antimicrobial drugs. Salmonella is generally isolated less than 1% of the time from
the feces of dogs and cats that are fed processed commercial

Etiology and Epidemiology


TABLE 45-1
Salmonella are gram-negative, motile, non–spore-forming fac- Classification of Salmonellae
ultative anaerobic rods that belong to the family Enterobacte-
riaceae. Most Salmonella exist in two phases, which differ in Salmonella Salmonella
composition of their flagellar antigens. Species ­Subspecies Example Serotypes
Salmonella are ubiquitous organisms that can be isolated
from the intestinal tracts of an enormous variety of animal spe- Salmonella enterica (I) Salmonella ser. Typhimurium
cies and are a major cause of enterocolitis and sometimes severe enterica S. ser. Typhi
systemic illness in humans and animals. Infections can be trans- S. ser. Choleraesuis
ferred between animal species and are zoonotic. Many infec- salamae (II) S. ser. Greenside
tions are food borne, especially in association with milk, meat, arizonae (IIIa) S. IIIa 18:z4,z23:—
and eggs. Salmonella spp. have a remarkable ability to survive
diarizonae (IIIb) S. IIIb 60:k:z
long periods of time (weeks to years) in the environment, and
so fomite transmission is important. In addition, Salmonella can houtenae (IV) S. ser. Marina
multiply rapidly in contaminated food left at room temperature indica (VI) S. ser. Srinagar
and can survive freezing for several weeks.2 Salmonella V S. ser. Brookfield
The genus Salmonella comprises two species, Salmonella bongori
enterica and Salmonella bongori, each of which contains

437
438 SECTION 2  Bacterial Diseases

pet foods,15-19 although one study of household pet dogs in tissues, and intestinal epithelium, along with evasion of the host
Ontario, Canada revealed a shedding prevalence of 23%.20 immune response, is followed by persistent shedding, often for
The highest rates of infection have been found in group-housed several weeks, after which animals can become latently infected.
dogs, especially those fed raw meat diets, such as racing sled Reactivation of shedding, with or without clinical illness, may
dogs and greyhound breeding facilities, where prevalences have occur, sometimes as a result of stress or immunosuppression.27,28
exceeded 75%.8,21 Salmonella was isolated from the feces of 18 If the organism has properties that allow it to spread systemi-
of 26 (69%) healthy pre-race Alaskan sled dogs, and 19 of 30 cally and host defenses are impaired, bacteremia and extraint-
(63%) diarrheic racing Alaskan sled dogs, which underscored estinal infection follows.29 More virulent strains have a greater
the lack of an association between the isolation of Salmonella ability to multiply intracellularly in nonphagocytic cells. Fever,
and clinical diarrhea.8 Coprophagic behavior may also contrib- hypoglycemia, and leukopenia occur. The Salmonella lipo-
ute to salmonellosis in dogs. In cats, the feeding of raw poultry polysaccharide (LPS) can then induce endotoxic shock, with
may increase risk for Salmonella infection,22 and occasionally, hypotension and activation of the complement and coagulation
high prevalences of infection have been detected in group- cascade. The coagulation cascade is also activated with exposure
housed cats18 and shelter cats. Ingestion of infected wild bird to Salmonella porins, which are hydrophobic outer membrane
species by cats during seasonal bird migrations can also lead proteins. Disseminated intravascular coagulation (DIC) follows.
to salmonellosis (songbird fever). In a study of diarrheic and Dogs and cats infected with Salmonella spp. may show no signs
nondiarrheic shelter cats in Florida, the prevalence of Salmo- or they may develop enterocolitis, focal suppurative infection, or
nella shedding as determined by PCR assay was 6% and 4%, severe systemic illness. The majority of dogs are chronically and
respectively.23 Susceptibility to shedding and disease is highest subclinically infected. When disease occurs, signs often begin 3 to
in immunosuppressed dogs and cats, including young animals, 5 days after infection or onset of immunosuppression. Fever (occa-
pregnant animals, those in overcrowded conditions, and those sionally as high as 106°F [41°C]), lethargy, and anorexia may be
with underlying immunosuppressive illness (such as neoplasia, followed by abdominal pain, vomiting, and watery to mucoid,
diabetes mellitus, retroviral infection, and immune-mediated often hemorrhagic diarrhea, and dehydration. Weight loss may be
disease) or immunosuppressive drug therapy.24,25 seen. Diarrhea may take several weeks to resolve. Rarely, chronic
intermittent diarrhea that lasts up to 8 weeks develops.30
Clinical Features Severely affected animals develop signs of septic shock (see
Chapter 86). Neurologic signs, and signs that relate to endocar-
Signs and Their Pathogenesis ditis, arthritis, pancreatitis, pneumonia, peritonitis, and chole-
After ingestion, salmonellae that survive the acidic environment cystitis occur in some animals. Infected cats may develop severe
of the stomach gain access to the intestine, where they disrupt enterocolitis, with fever, pancytopenia, and hyperbilirubinemia.
tight junctions and actively invade (1) enterocytes; (2) M cells Some cats show persistent fever and anorexia with no diarrhea.
within the ileum (which sample luminal antigens); and (3) den- Conjunctivitis has been described in cats in association with Sal-
dritic cells, which send protrusions into the gut lumen between monella infection.31
intestinal epithelial cells. Interaction of Salmonella with immune Occasionally salmonellae localize in a particular organ, such
cells and epithelial cells within the gut leads to production of as the lungs or urinary tract (Figure 45-1). Signs of dysfunction
cytokines and chemokines, with massive influx of lymphocytes, of that organ system may follow, even when enteric signs or
macrophages, and neutrophils, sometimes with severe epithe- positive fecal culture results are absent.32 Abortion and stillbirth
lial injury and mucosal sloughing.26 Localization in the mesen- can occur after transplacental infection, and the dam may also
teric lymph nodes, Peyer’s patches, solitary intestinal lymphoid infect her neonates, which leads to fading puppies.33-35

FIGURE 45-1  Large, chronic pancreatic abscess due to Salmonella Typhimurium associated with a pancreatic adenocarcinoma in a 15-year-old castrated male domestic shorthair
cat. The mass was multicystic and fluctuant, and when cut open was filled with cloudy fluid and lined by thick tan debris. (Courtesy University of California, Davis, Veterinary Anatomic
­Pathology Service.)
CHAPTER 45  Salmonellosis 439

Diagnosis lymphadenomegaly, intestinal wall thickening, fluid-filled bowel


segments, ­hepatomegaly, splenomegaly, or ascites.
The traditional diagnosis of canine and feline salmonellosis is
made based on culture of the organism in conjunction with Microbiologic Testing
clinical signs and assessment of potential risk factors, such as Bacterial Isolation
hospitalization, age, environmental exposure, and antibiotic A diagnosis of salmonellosis can be made when Salmonella is
administration. Diagnostic assays available for salmonellosis in isolated from a normally sterile site, such as the blood, bron-
dogs and cats are shown in Table 45-2. choalveolar lavage specimens, synovial fluid, or urine specimens
collected by cystocentesis. Isolation of Salmonella from feces
Laboratory Abnormalities does not confirm that the organism is the cause of disease, but
Complete Blood Count it can raise suspicion that Salmonella may be playing a role in
The CBC in dogs or cats with salmonellosis may be unremark- enterocolitis, and it has zoonotic significance (see Public Health
able or may show a neutrophilia with a left shift; in bacteremic Aspects, later). Isolation also allows antimicrobial susceptibility
animals, anemia, a degenerative left shift, toxic neutrophils, testing, which is important because many isolates of Salmonella
lymphopenia, and thrombocytopenia are usually present. are resistant to multiple antimicrobial drugs.
Rarely, intracellular bacteria are seen within neutrophils. Salmonella grow readily at 37°C on routine bacteriologic
media from specimens that have been collected from a nor-
Serum Chemistry Profile mally sterile site. Selective media are required for specimens
In animals with severe salmonellosis, changes on the serum that are heavily contaminated with other bacteria such as feces.
chemistry profile include elevated liver enzyme activities, hyper- Examples of selective media include enrichment broths such
bilirubinemia, azotemia, hypoalbuminemia, hypocholesterol- as tetrathionate broth or Selenite F broth. After enrichment,
emia, hypoglycemia, and electrolyte abnormalities. subculturing is performed on a medium such as deoxycho-
late, which also favors growth of Salmonella. Many veteri-
Urinalysis nary laboratories incorporate such protocols for isolation of
Salmonella spp. occasionally localize in the kidneys, with asso- Salmonella as part of a panel for detection of enteropatho-
ciated isosthenuria, pyuria, proteinuria, casts, and bacteriuria. genic bacteria. Once Salmonella is isolated, the organism can
be identified based on biochemical reactions and serotyping,
Clotting Function which involves agglutination testing with O and H antisera.
Dogs and cats that develop DIC may have clotting func- In the future, use of whole cell matrix-assisted laser desorp-
tion abnormalities that include prolonged coagulation times, tion ionization-time-of-flight (MALDI-TOF) mass spectrom-
increased fibrin degradation product concentrations, and etry in veterinary clinical microbiology laboratories should
decreased antithrombin concentrations. allow identification of Salmonella at the species and subspe-
cies level within minutes.36 It also can identify some important
Diagnostic Imaging S. enterica subsp. enterica to the serovar level, and could
Plain Radiography reduce the need for laborious subtyping procedures.37
Thoracic radiographs in animals with salmonellosis may reveal
bronchointerstitial to alveolar infiltrates in animals with pneu- Molecular Diagnosis Using the Polymerase Chain Reaction
monia, or pleural effusion with pyothorax. Abdominal radio- Some veterinary diagnostic laboratories offer PCR assays for
graphs can reveal fluid-filled intestines, decreased serosal detail, Salmonella, some of which have been shown as highly sensi-
and/or mild hepatomegaly. tive and specific when compared with culture.38,39 PCR is more
rapid than culture but currently does not provide information
Sonographic Findings on antimicrobial susceptibility. It has been recommended that
Depending on the severity of the infection, abdominal PCR assay after overnight enrichment in a nonselective broth be
ultrasound may be unremarkable in dogs and cats with adopted as the gold standard for diagnosis, and that all speci-
salmonellosis, or show changes that include abdominal mens that test positive by PCR assay be cultured using selective

TABLE 45-2
Diagnostic Assays Available for Salmonellosis in Dogs and Cats
Assay Specimen Type Target Performance
Bacterial Feces, blood, synovial fluid, tis- Salmonella species Sensitive and specific. False negatives may occur, especially
isolation sue aspirates, bronchoalveolar following antimicrobial therapy. Isolation from feces
­lavage fluid, peritoneal or pleu- does not imply that Salmonella is the cause of disease.
ral effusions, urine, CSF, tissues Allows subsequent antimicrobial susceptibility testing.
obtained at necropsy
PCR As for isolation Salmonella DNA Rapid (within hours); can be highly sensitive and specific.
Positive results from feces do not imply that Salmonella
is the cause of disease. Does not allow susceptibility test-
ing or typing for epidemiologic purposes.
440 SECTION 2  Bacterial Diseases

enrichment to isolate and identify the infecting organism.40 As Dogs and cats with systemic salmonellosis may require
with culture, detection of the organism in feces using PCR assays aggressive intravenous fluid therapy and colloidal support with
does not prove that it is the cause of gastrointestinal illness. hydroxyethyl starch or plasma, together with parenteral anti-
microbial drugs. The choice of antimicrobial drugs should be
Pathologic Findings based on the results of culture and susceptibility, because of the
Gross Pathologic Findings potential for resistance to multiple antimicrobial drugs through
Gross necropsy findings in dogs and cats with severe salmo- plasmid transfer. In the event of systemic disease, administra-
nellosis include widespread petechial and ecchymotic hemor- tion of a combination of ampicillin and a fluoroquinolone is
rhages, hemorrhagic enteritis, abscesses within parenchymal advocated as empiric therapy while awaiting results of culture
organs, enlarged mesenteric lymph nodes, and fibrinohemor- and susceptibility. Because of the importance of endotoxin in
rhagic ascites fluid. Pulmonary consolidation and edema may the pathogenesis of sepsis due to gram-negative bacteria such
also be present. as Salmonella, considerable effort has been invested in devel-
opment and application of drugs targeting endotoxin.43 Clear
Histopathologic Findings benefit through reduction in mortality has not yet been dem-
Histopathologic lesions in dogs and cats with salmonellosis onstrated for these treatments, and to date they have not been
are highly variable. Changes in severely affected animals may widely used in dogs and cats. Most recently, investigations have
include suppurative pneumonia, necrotizing hepatitis, and nec- been focused on antagonists of Toll-like receptor 4, which is the
rotizing and fibrinohemorrhagic enterocolitis, typhlitis, and LPS receptor.44 Eritoran tetrasodium (E5564) and TAK-242 are
cholecystitis. More chronic lesions such as chronic cholecys- examples of TLR4 antagonists that have been used in clinical
titis have also been described.41,42 Sometimes, gram-negative trials involving septic humans.45
bacilli are identified within lesions with tissue Gram stains
(Figure 45-2). Immunity and Vaccination
Treatment and Prognosis Salmonella spp. can persist in the host by interfering with den-
dritic cell function in the intestinal tract. The organism pene-
The movement of dogs and cats that test positive for Salmonella trates dendritic cells and avoids lysosomal degradation, which
should be limited, and if possible, hospitalized animals should prevents subsequent antigen presentation by dendritic cells on
be isolated. The mere detection of Salmonella in the feces of molecules of the major histocompatibility complex.46 Reduced
dogs and cats with uncomplicated diarrhea does not warrant intracellular proliferation of the organism within antigen-
antimicrobial administration, and supportive care only is rec- presenting cells may also limit antigen presentation and the
ommended. Most of these animals have self-limiting disease and development of an effective immune response.47 Although Sal-
shedding, and injudicious antimicrobial administration has the monella vaccines have been developed, none are available for
potential to prolong the carrier state and contribute to antimi- dogs and cats.
crobial resistance. Treatment of an animal with uncomplicated
diarrhea is also not advocated if the owner is immunocompro- Prevention of Enteric Bacterial Infections
mised, although appropriate husbandry recommendations and
barrier control must be enforced. Prevention of enteric bacterial infections includes feeding dogs
and cats properly cooked foods, hand washing before and after
handling pets and pet food, and prevention of coprophagic
behavior. Proper handling and storage of pet foods can also
limit the potential for food contamination during storage by
rodents, insects, and human hands.
Within the hospital setting, fomites such as food dishes,
endoscopes, proctoscopes, and rectal thermometers should be
disinfected between uses, and single-use disposable thermom-
eter covers should be used. Cages should be cleaned and prop-
erly disinfected (see Chapter 11) between animals. Blood donors
should not be housed with the regular hospital population, as
they may be a source of Salmonella infection.
Dogs and cats diagnosed with salmonellosis or Salmonella
shedding should be isolated from other animals until shedding
stops. Fecal cultures should be repeated every 2 weeks, and ter-
mination of shedding is identified when three successive nega-
tive cultures have been obtained.

Public Health Aspects


All enteric bacterial infections of dogs and cats have the poten-
FIGURE 45-2  Gram-negative bacilli (arrow) in the lungs of a cat with suppurative tial to infect and cause illness in humans. Nontyphoid salmo-
cholecystitis and pneumonia due to Salmonella arizonae. The cat had been treated with nellosis (salmonellosis other than that caused by S. ser. Typhi)
prednisone and cyclosporine for immune-mediated hemolytic anemia. Brown and Benn can be transmitted from domestic animals to humans and
stain, 1000× magnification. cause serious enterocolitis, with fever, severe abdominal pain,
CHAPTER 45  Salmonellosis 441

diarrhea, nausea, inappetence, chills, and headache. Serious been isolated from dogs, cats, and reptiles.20,48 People exposed
complications tend to occur in children, the elderly, or other- to dogs and cats that are fed raw meat diets are at increased
wise immunocompromised individuals such as those infected risk of exposure.
with HIV. The majority of cases result from ingestion of Proper hand washing after handling pets, pet food, and fomi-
improperly cooked foods of animal origin, such as meat, milk, tes such as bedding and food dishes can help prevent zoonotic
and eggs. Most, if not all, pet reptiles carry Salmonella, and transmission of Salmonella. Reptile ownership by families with
there has been considerable effort to educate the public about children younger than 5 years of age or among the immuno-
the hazards of reptile-associated salmonellosis. Dogs, and to a suppressed is discouraged by public health authorities.49 Proper
lesser extent cats, are less commonly incriminated as a source of food handling practices, including separating raw meat from
Salmonella. Of particular concern, multidrug-resistant Salmo- vegetables and proper cooking of meat, can also reduce salmo-
nella strains, such as S. ser. Typhimurium strain DT 104, have nellosis in humans.

CASE EXAMPLE thorax 3 weeks before referral that was assumed to be a foxtail
abscess. Sampson had a good appetite. Increased thirst and
urination had been noted after the prednisone treatment was
initiated. There had been no coughing, sneezing, vomiting, or
diarrhea. He was fed a duck- and potato-based diet for food
allergy dermatitis and had been treated intermittently with
prednisone, 4 mg PO q24h, for this condition since he was 6
months old. He had not traveled outside his local area, but
he lived on a ranch and was known to consume horse feces
and dead wild animals. There was no known tick exposure. He
was up to date on vaccinations including canine distemper,
adenovirus, parvovirus, and rabies vaccines.
Current medications: Prednisone 0.9 mg/kg PO q12h
Physical Examination:
Body Weight: 35.8 kg
General: Quiet, alert and responsive, 5% to 7% dehydrated.
Recumbent. T = 102.9°F (39.4°C), HR = 140 beats/min,
panting, mucous membranes pink and tacky, CRT = 1 s.
Integument: Multifocal epidermal collarettes were present
and the haircoat was dry. A shaved area was present on the
left lateral thoracic wall.
Eyes, Ears, Nose, and Throat: Moderate periodontal disease was
present. A fundic examination was within normal limits.
Musculoskeletal: Body condition score was 5/9 with pelvic
limb muscle atrophy. Pain and effusion of the carpi, tarsi,
and stifles with decreased range of motion was noted.
Spinal and pericervical pain was detected on palpation of
these regions.
Cardiovascular: Weak but synchronous femoral pulses were
noted. No murmurs or arrhythmias were auscultated.
FIGURE 45-3  “Sampson”, a 3-year-old male castrated shepherd mix with Respiratory: There were moderately increased breath sounds
salmonellosis. in all lung fields.
Gastrointestinal/Genitourinary: The abdomen was tense
Signalment: “Sampson,” a 3-year-old male castrated shepherd on palpation and moderate hepatomegaly was detected.
mix from Fresno in southern California (Figure 45-3). The dog urinated a large amount of odiferous urine during
History: Sampson was brought to a veterinarian for evaluation the examination. No abnormalities were detected on rectal
of a 3-month history of bilateral tarsal joint swelling. Some examination.
clinical improvement had been noted after treatment with Lymph Nodes: Moderate peripheral lymphadenopathy was
oral tetracycline (28 mg/kg PO q8h) for 2 weeks. Subsequently, present. Mandibular, superficial cervical, and popliteal
he was treated with oral prednisone (1.1 mg/kg PO q12h). Two lymph nodes were all 1.5 to 2.5 cm in diameter.
weeks after the prednisone treatment was initiated, Sampson Laboratory Findings:
became nonambulatory in his pelvic limbs, and all of his CBC:
peripheral joints became swollen and he was referred to the HCT 35.4% (40%-55%)
University of California, Davis, Veterinary Medical Teaching MCV 67 fL (65-75 fL)
Hospital. A mass had been removed from his left dorsolateral MCHC 35.6 g/dL (33-36 g/dL)

Continued
442 SECTION 2  Bacterial Diseases

Reticulocytes 101,200 cells/µL (7000-65,000 cells/µL)


WBC 7870 cells/µL (6000-13,000 cells/µL)
Neutrophils 6139 cells/µL (3000-10,500 cells/µL)
Band neutrophils 1495 cells/µL
Metamyelocytes 79 cells/µL
Lymphocytes 0 cells/µL (1000-4000 cells/µL)
Monocytes 79 cells/µL (150-1200 cells/µL)
Platelets 419,000 platelets/µL (150,000-400,000 platelets/µL).
Neutrophils, band neutrophils, and metamyelocytes all
showed moderate toxic changes.
Serum Chemistry Profile:
Sodium 148 mmol/L (145-154 mmol/L)
Potassium 4.4 mmol/L (3.6-5.3 mmol/L)
Chloride 105 mmol/L (108-118 mmol/L)
Bicarbonate 14 mmol/L (16-26 mmol/L)
Phosphorus 5.8 mg/dL (3.0-6.2 mg/dL)
Calcium 10.3 mg/dL (9.7-11.5 mg/dl)
BUN 10 mg/dL (5-21 mg/dL)
Creatinine 0.4 mg/dL (0.3-1.2 mg/dL)
Glucose 111 mg/dL (64-123 mg/dL)
FIGURE 45-4  Thickening and hyperechogenicity of the posterior tricuspid valve
Total protein 6.1 g/dL (5.4-7.6 g/dL)
­leaflet (arrowhead) in a dog with Salmonella arizonae bacteremia.
Albumin 2.3 g/dL (3.0-4.4 g/dL)
globulin 3.8 g/dL (1.8-3.9 g/dL)
ALT 560 U/L (19-67 U/L) IFA for antibodies to Bartonella vinsonii subspecies berkhoffii
AST 72 U/L (19-42 U/L) was negative. Serology for Coccidioides spp. antibodies was
ALP 3318 U/L (21-170 U/L) also negative.
Creatine kinase 197 U/L (51-399 U/L) Aerobic bacterial culture of blood, joint fluid, and urine collect-
Gamma GT 59 U/L (0-6 U/L) ed by cystocentesis: Salmonella arizonae, susceptible to all
Cholesterol 212 mg/dL (135-361 mg/dL) antimicrobials tested.
Total bilirubin 0.3 mg/dL (0-0.2 mg/dL). Diagnosis: Salmonella arizonae septic polyarthritis,
Urinalysis: SGr 1.010; pH 8.0, 1+ protein (SSA), 3+ hemoprotein, bacteremia, and possible tricuspid valve endocarditis.
no bilirubin or glucose, 25-35 WBC/HPF, 10-15 RBC/HPF, Treatment: Before obtaining susceptibility results, Sampson
many rods. was treated with intravenous crystalloids, opioids for pain
Imaging Findings: control, enrofloxacin (5 mg/kg IV q24h), and ampicillin (30
Thoracic Radiographs: The cardiovascular and pulmonary mg/kg IV q8h). The ampicillin was discontinued when the
structures appeared within normal limits. susceptibility results became available. The prednisone was
Right and Left Carpal, Stifle, and Tarsal Radiographs: tapered over a 3-day period and discontinued. The dog was
Intracapsular soft tissue swelling was associated with all moved to an isolation ward on diagnosis of salmonellosis.
joints. During hospitalization, Sampson’s appetite declined initially,
Spinal Radiographs: No abnormalities were detected. and he remained very painful for 6 days. After that time, he
Abdominal Ultrasound: The liver was enlarged and was able to sit up, began eating, and was discharged 8 days
hyperechoic. There was echogenic sediment in the urinary after admission.
bladder. Comments: Salmonella arizonae infections in humans have
Echocardiogram: Thickening and hyperechogenicity of the been associated with direct or indirect contact with reptiles.
posterior leaflet of the tricuspid valve was noted (arrowhead) Reptile-associated salmonellosis is an increasing problem
which raised suspicion for endocarditis (Figure 45-4). in humans in the United States and is especially a problem
Cytology Findings: Arthrocentesis of right and left carpi, in infants and immunocompromised adults in contact with
right tarsus and right stifle: Purulent inflammation was snakes and iguanas, or those who eat sun-dried rattlesnake
noted in the right tarsus, right stifle, and left carpus, with meat for medicinal purposes. Horse feces from the farm were
approximately 85% neutrophils and the remaining cells cultured for Salmonella and were negative. It is possible that
were a mixture of small and large mononuclear cells. In the infection in this dog occurred as a result of ingestion
the right stifle, degenerate neutrophils are visualized, and of wild reptiles. The long history of glucocorticoid therapy
some contained intracellular rod-shaped bacteria. for atopic dermatitis in this dog may have contributed
Microbiologic Testing: Serologic testing: An in-clinic to immunosuppression and predisposition to systemic
ELISA for antibodies to Borrelia burgdorferi, Anaplasma salmonellosis.
phagocytophilum, and Ehrlichia canis was negative. Serum
CHAPTER 45  Salmonellosis 443

SUGGESTED READINGS 19. Murphy C, Reid-Smith RJ, Prescott JF, et al. Occurrence of antimi-
crobial resistant bacteria in healthy dogs and cats presented to pri-
Behravesh CB, Ferraro A, Deasy M, et al. Human Salmonella infections vate veterinary hospitals in southern Ontario: a preliminary study.
linked to contaminated dry dog and cat food, 2006-2008. Pediatrics. Can Vet J. 2009;50(10):1047-1053.
2010;126(3):477-483. 20. Leonard EK, Pearl DL, Finley RL, et  al. Comparison of antimi-
Grassl GA, Finlay BB. Pathogenesis of enteric Salmonella infections. crobial resistance patterns of Salmonella spp. and Escherichia coli
Curr Opin Gastroenterol. 2008;24(1):22-26. recovered from pet dogs from volunteer households in Ontario
Morley PS, Strohmeyer RA, Tankson JD, et  al. Evaluation of the (2005-2006). J Antimicrob Chemother. 2012;67:174-181.
association between feeding raw meat and Salmonella enterica 21. Morley PS, Strohmeyer RA, Tankson JD, et al. Evaluation of the
infections at a Greyhound breeding facility. J Am Vet Med Assoc. association between feeding raw meat and Salmonella enterica
2006;228(10):1524-1532. infections at a Greyhound breeding facility. J Am Vet Med Assoc.
2006;228(10):1524-1532.
REFERENCES 22. Philbey AW, Brown FM, Mather HA, et al. Salmonellosis in cats in
1. Salmon DE, Smith T. Investigations in swine plague. USDA Bur the United Kingdom: 1955 to 2007. Vet Rec. 2009;164(4):120-122.
Anim Indus Ann Rep. 1885:184. 23. Sabshin SJ, Levy JK, Tupler T, et al. Enteropathogens identified in
2. Dominguez SA, Schaffner DW. Survival of salmonella in pro- cats entering a Florida animal shelter with normal feces or diarrhea.
cessed chicken products during frozen storage. J Food Prot. J Am Vet Med Assoc. 2012;241:331-337.
2009;72(10):2088-2092. 24. Hohenhaus AE, Rosenberg MP, Moroff SD. Concurrent lymphoma
3. Brenner FW, Villar RG, Angulo FJ, et al. Salmonella nomenclature. and salmonellosis in a cat. Can Vet J. 1990;31(1):38-40.
J Clin Microbiol. 2000;38(7):2465-2467. 25. Moazed TC, Deeb BJ, DiGiacomo RF. Subcutaneous abscess due to
4. Foti M, Daidone A, Aleo A, et al. Salmonella bongori 48:z35:— in Salmonella adelaide in a grey collie with cyclic hematopoiesis. Lab
migratory birds, Italy. Emerg Infect Dis. 2009;15(3):502-503. Anim Sci. 1990;40(6):639-641.
5. Finley R, Ribble C, Aramini J, et al. The risk of salmonellae shed- 26. Grassl GA, Finlay BB. Pathogenesis of enteric Salmonella infec-
ding by dogs fed Salmonella-contaminated commercial raw food tions. Curr Opin Gastroenterol. 2008;24(1):22-26.
diets. Can Vet J. 2000;48(1):69-75. 27. Delaloye J, Merlani G, Petignat C, et  al. Nosocomial nontyphoi-
6. Lefebvre SL, Reid-Smith R, Boerlin P, et al. Evaluation of the risks dal salmonellosis after antineoplastic chemotherapy: reactivation
of shedding salmonellae and other potential pathogens by therapy of asymptomatic colonization? Eur J Clin Microbiol Infect Dis.
dogs fed raw diets in Ontario and Alberta. Zoonoses Public Health. 2004;23(10):751-758.
2008;55(8-10):470-480. 28. Lomborg SR, Agerholm JS, Jensen AL, et al. Effects of experimen-
7. Lenz J, Joffe D, Kauffman M, et al. Perceptions, practices, and con- tal immunosuppression in cattle with persistently high antibody
sequences associated with foodborne pathogens and the feeding of levels to Salmonella dublin lipopolysaccharide O-antigens. BMC
raw meat to dogs. Can Vet J. 2009;50(6):637-643. Vet Res. 2007;3:17.
8. McKenzie E, Riehl J, Banse H, et  al. Prevalence of diarrhea 29. Fierer J, Guiney DG. Diverse virulence traits underlying dif-
and enteropathogens in racing sled dogs. J Vet Intern Med. ferent clinical outcomes of Salmonella infection. J Clin Invest.
2010;24(1):97-103. 2001;107(7):775-780.
9. Finley R, Reid-Smith R, Ribble C, et al. The occurrence and anti- 30. Dow SW, Jones RL, Henik RA, et  al. Clinical features of sal-
microbial susceptibility of salmonellae isolated from commercially monellosis in cats: six cases (1981-1986). J Am Vet Med Assoc.
available canine raw food diets in three Canadian cities. Zoonoses 1989;194(10):1464-1466.
Public Health. 2008;55(8-10):462-469. 31. Fox JG, Beaucage CM, Murphy JC, et al. Experimental S­ almonella-
10. Centers for Disease Control and Prevention. Multistate outbreak associated conjunctivitis in cats. Can J Comp Med. 1984;48(1):
of human Salmonella infections caused by contaminated dry dog 87-91.
food: United States, 2006-2007. MMWR Morb Mortal Wkly Rep. 32. Rodriguez Jr CO, Moon ML, Leib MS. Salmonella choleraesuis
2008;57(19):521-524. pneumonia in a cat without signs of gastrointestinal disease. J Am
11. Behravesh CB, Ferraro A, Deasy M, et  al. Human Salmonella Vet Med Assoc. 1993;202(6):953-955.
infections linked to contaminated dry dog and cat food, 2006- 33. Morse EV, Duncan MA. Canine salmonellosis: prevalence, epizoot-
2008. Pediatrics. 2010;126(3):477-483. iology, signs, and public health significance. J Am Vet Med Assoc.
12. Selmi M, Stefanelli S, Bilei S, et al. Contaminated commercial dehy- 1975;167(9):817-820.
drated food as source of multiple Salmonella serotypes outbreak in 34. Caldow GL, Graham MM. Abortion in foxhounds and a ewe
a municipal kennel in Tuscany. Vet Ital. 2011;47(2):183-190. flock associated with Salmonella montevideo infection. Vet Rec.
13. Centers for Disease Control and Prevention. Notes from the field: 1998;142(6):138-139.
human Salmonella infantis infections linked to dry dog food— 35. Redwood DW, Bell DA. Salmonella panama: isolation from
United States and Canada. MMWR Morb Mortal Wkly Rep. aborted and newborn canine fetuses. Vet Rec. 1983;112(15):362.
2012;2012(61):436. 36. Dieckmann R, Helmuth R, Erhard M, et  al. Rapid classification
14. Morse EV, Duncan MA, Estep DA, et al. Canine salmonellosis: a and identification of salmonellae by whole-cell matrix-assisted laser
review and report of dog to child transmission of Salmonella enter- desorption ionization-time of flight mass spectrometry. Appl Envi-
itidis. Am J Public Health. 1976;66(1):82-84. ron Microbiol. 2008;74:7767-7778.
15. Hill SL, Cheney JM, Taton-Allen GF, et  al. Prevalence of 37. Dieckmann R, Malorny B. Rapid screening of epidemiologically
enteric zoonotic organisms in cats. J Am Vet Med Assoc. important Salmonella enterica subsp. enterica serovars by whole-
2000;216(5):687-692. cell matrix-assisted laser desorption ionization-time of flight mass
16. Spain CV, Scarlett JM, Wade SE, et al. Prevalence of enteric zoo- spectrometry. Appl Environ Microbiol. 2011;77:4136-4146.
notic organisms in cats less than 1 year old in central New York 38. Kurowski PB, Traub-Dargatz JL, Morley PS, et  al. Detection of
State. J Vet Intern Med. 2001;15(1):33-38. Salmonella spp. In fecal specimens by use of real-time polymerase
17. Stavisky J, Radford AD, Gaskell R, et al. A case-control study of chain reaction assay. Am J Vet Res. 2002;63(9):1265-1268.
pathogen and lifestyle risk factors for diarrhea in dogs. Prev Vet 39. Pusterla N, Byrne BA, Hodzic E, et al. Use of quantitative real-time
Med. 2011;99:185-192. PCR for the detection of Salmonella spp. in fecal samples from horses
18. Van Immerseel F, Pasmans F, De Buck J, et al. Cats as a risk for at a veterinary teaching hospital. Vet J. 2010;186(2):252-255.
transmission of antimicrobial drug-resistant Salmonella. Emerg 40. Ward MP, Alinovi CA, Couëtil LL, Wu CC. Evaluation of a PCR
Infect Dis. 2004;10(12):2169-2174. to detect Salmonella in fecal samples of horses admitted to a veteri-
nary teaching hospital. J Vet Diagn Invest. 2005;17:118-123.
444 SECTION 2  Bacterial Diseases

41. Timbs DV, Durham PJ, Barnsley DJ. Chronic cholecystitis 46. Bueno SM, Gonzáles PA, Carreño LJ, et al. The capacity of Salmo-
in a dog infected with Salmonella typhimurium. N Z Vet J. nella to survive inside dendritic cells and prevent antigen presenta-
1974;22(6):100-102. tion to T cells is host specific. Immunology. 2008;124(4):522-533.
42. Stiver SL, Frazier KS, Mauei MJ, et  al. Septicemic salmonel- 47. Albaghdadi H, Robinson N, Finlay B, et al. Selectively reduced intra-
losis in two cats fed a raw-meat diet. J Am Anim Hosp Assoc. cellular proliferation of Salmonella enterica serovar typhimurium
2003;39(6):538-542. within APCs limits antigen presentation and development of a
43. Opal SM, Glück T. Endotoxin as a drug target. Crit Care Med. rapid CD8 T cell response. J Immunol. 2009;183(6):3778-3787.
2003;31(suppl 1):S57-S64. 48. Wright JG, Tengelsen LA, Smith KE, et  al. Multidrug-resistant
44. Leon CG, Tory R, Jia J, et al. Discovery and development of Toll- Salmonella Typhimurium in four animal facilities. Emerg Infect Dis.
like receptor 4 (TLR4) antagonists: a new paradigm for treating 2005;11(8):1235-1241.
sepsis and other diseases. Pharm Res. 2008;25(8):1751-1761. 49. Centers for Disease Control and Prevention. http://www.cdc.gov/
45. Wittebole X, Castanares-Zapatero D, Laterre PF. Toll-like ­receptor Features/salmonellafrogturtle. Last accessed December 23, 2012.
4 modulation as a strategy to treat sepsis. Mediators Inflamm.
2010;2010:568396.
CHAPTER 46

Enteric Escherichia coli Infections


Jane E. Sykes and Stanley L. Marks

Each pathovar is defined by a characteristic set of virulence fac-


Overview of Enteric Escherichia coli Infections tors that act in concert to determine the clinical, pathologic, and
First Described: Germany, 1885 (Theodor Escherich)1 epidemiologic features of the disease they cause. The seven path-
Cause: Escherichia coli (gram-negative rods that belong to the ovars include enteropathogenic E. coli (EPEC), enterotoxigenic
Enterobacteriaceae) E. coli (ETEC), enterohemorrhagic E. coli (EHEC), necrotoxi-
genic E. coli (NTEC), enteroinvasive E. coli (EIEC), enteroag-
Affected Hosts: Humans and a large variety of animals
gregative E. coli (EAEC), and adherent-invasive E. coli (AIEC)
Geographic Distribution: Worldwide (Table 46-1). Currently, the epidemiology of many of these strains
Primary Mode of Transmission: Fecal-oral and their role in disease causation is not well defined in small
animals, with most work having been done in humans and farm
Major Clinical Signs: Fever, lethargy, inappetence, diarrhea, animal species. Many strains have been isolated from both nondi-
vomiting arrheic and diarrheic dogs and cats. An association between EPEC
Differential Diagnoses: Differential diagnoses for suspected and EHEC (including H7:O157) infection and diarrhea in dogs
E. coli ­enterocolitis include canine and feline parvovi- has been detected.2,3 In addition, AIEC strains have been well
rus infection, canine distemper virus infection, salmon associated with granulomatous colitis in boxer dogs.4
poisoning disease, campylobacteriosis, clostridial
diarrhea, salmonellosis, giardiasis, tritrichomoniasis Clinical Features
(cats), cryptosporidiosis, whipworms, pythiosis, histo-
plasmosis, leptospirosis, dietary indiscretion, gastroin- Signs and Their Pathogenesis
testinal foreign body, pancreatitis, inflammatory bowel EPEC strains carry the eaeA gene on their chromosome (E. coli
disease, lymphoma, hypoadrenocorticism, hyperthyroid- attaching effacing), which is located in a “pathogenicity island”
ism (cats), toxins (including drugs). referred to as LEE (locus of enterocyte effacement). The eaeA
Human Health Significance: Dogs and cats may be a source of gene encodes a 94-kDa protein, intimin, which allows the organ-
human infection with some E. coli serotypes. ism to adhere intimately to intestinal epithelial cells. This leads
to local effacement of the microvilli and formation of numerous,
actin-rich pedestals on which the bacteria reside (Figure 46-1).5
The bacteria secrete their own receptor, the Tir receptor, and
Etiology and Epidemiology Tir-intimin interactions are involved in pedestal formation.
The pedestals may help the bacteria to remain extracellular,
Escherichia coli are pleomorphic gram-negative, non–spore- and thus escape immune recognition by the host. The result-
forming rods that belong to the family Enterobacteriaceae. Like ing characteristic histopathologic lesions on enterocytes have
Salmonella, E. coli can survive long periods of time in feces, been referred to as “attaching and effacing lesions.” Subsequent
dust, and water. They are part of the normal flora of the gas- signal transduction events induced by EPEC may be associated
trointestinal tract but can be associated with enterocolitis in the with loss of tight junction integrity and altered electrolyte trans-
presence of bacterial virulence factors and impaired local or port, with associated watery diarrhea. The EPEC pathovar does
systemic immunity. As with Salmonella infections, E. coli infec- not produce Shiga toxin, Shiga-like toxin, or verotoxins. EPEC
tions can be transferred between animal species and some may serotypes that have been associated with diarrhea in humans
be zoonotic; many infections are food borne. have been identified in dogs and cats.6,7
There are more than 170 serogroups of E. coli based on the ETEC are a major cause of diarrhea in human infants in
identity of the bacterial O (somatic) antigen, the sugar that is developing countries and are the agents most frequently respon-
on the most external portion of the bacterial lipopolysaccha- sive for traveler’s diarrhea. ETEC pathovars have been associ-
ride (LPS) (see Chapter 36). Organisms are also identified on ated with up to 31% of cases of canine diarrhea, particularly
the basis of their H (flagellar) antigens—for example, O157:H7 in young dogs.8-10 Most of the canine ETEC strains express ST
E. coli. Both the bacterial LPS (endotoxin) and the flagellar anti- enterotoxin, whereas ETEC strains that produce LT enterotoxin
gens are bacterial virulence factors. Other virulence factors in are rarely found. The bacteria adhere to the proximal small
E. coli include adhesins, bacterial exotoxins such as cytotoxic intestinal mucosa and produce plasmid-encoded LT and ST
necrotizing factor (CNF), hemolysins, heat-labile (LT) and heat- enterotoxins. LT is related to cholera toxin and is taken up by
stable (ST) enterotoxins, and the capsular polysaccharide (K). enterocytes. Once within intestinal epithelial cells, it activates
E. coli strains that cause gastrointestinal disease have been adenylate cyclase, which leads to increased concentrations of
divided into seven distinct pathogenic categories (pathovars). intracellular cyclic AMP with resultant secretion of electrolytes

445
446 SECTION 2  Bacterial Diseases

TABLE 46-1
Pathogenic Strains of Escherichia coli That Cause Enterocolitis
Strain Lesion Signs Virulence Characteristic
Enteropathogenic E. coli Attaching and effacing lesions Watery diarrhea eaeA gene (encodes
(EPEC) (effacement of microvilli and intimin)
pedestal formation)
Enterotoxigenic E. coli Adhere using fimbria and Diarrhea LT and ST production
(ETEC) produce heat-labile (LT)
and/or heat-stable (ST) toxins
Enterohemorrhagic E. coli Produce Shiga-like toxins Diarrhea, hemorrhagic colitis, Shiga-like toxin
(EHEC); also known as ­(verotoxins) that cause hemolytic-uremic syndrome ­production, see also
Shiga-toxin–producing vascular endothelial damage, EPEC
E. coli (STEC) or verotoxin- also produce attaching and
producing E. coli (VTEC) effacing lesions
Necrotoxigenic E. coli Produce cytotoxic necrotizing Diarrhea, bacteremia, urinary CNFs
(NTEC) ­factors (CNFs) tract infections in humans
Enteroinvasive E. coli Actively invade colonic Large bowel diarrhea Plasmid-encoded
(EIEC) epithelial cells ­invasion genes such as
ipaH
Adherent-invasive E. coli Invade colonic epithelial cells Granulomatous/histiocytic None known
(AIEC) using unknown virulence ulcerative colitis in dogs
mechanisms (especially boxers)
Enteroaggregative E. coli Adhere to intestinal epithelium Persistent watery diarrhea in aggR gene
(EAEC) via fimbriae and aggregate in humans
bricklike fashion

A B
FIGURE 46-1  Scanning and transmission electron micrographs showing actin pedestals induced by enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC).
A, EPEC generates attaching and effacing (AE) lesions on the intestinal epithelium after infection of gnotobiotic piglets. Note the pedestal-like structures on host cells beneath attached
bacteria. (From Campellone KG, Leong JM. Tails of two Tirs: actin pedestal formation by enteropathogenic E. coli and enterohemorrhagic E. coli. Curr Opin Microbiol 2003;6(1):82-90.)
B, Actin pedestals that resemble AE lesions formed in vivo are also generated on cultured epithelial (HeLa) cells. (Courtesy of Knutton S. In: Knutton S, Rosenshine L, Pallen, MJ, et al. A novel
EspA-associated surface organelle of enteropathogenic Escherichia coli involved in protein translocation into epithelial cells. EMBO J. 1998;17:2166-2176.)

and water that lead to diarrhea, hypovolemia, and metabolic They cause diarrhea, hemorrhagic colitis, and hemolytic-uremic
acidosis. LT may also downregulate innate host immune syndrome (HUS) (e.g., E. coli strain O157:H7). Shiga-like toxins
responses and promote ETEC adherence. In contrast, ST binds are absorbed from the intestinal lumen and cause vascular endo-
to the extracellular domain of guanylyl cyclase C, which leads thelial damage, which may lead to a multiorgan thrombotic pro-
to accumulation of intracellular cyclic GMP and ultimately cess in the absence of bacteremia. EHEC can also produce other
secretion of chloride and decreased absorption of NaCl, with toxins that contribute to the pathogenesis of the disease. HUS
resultant osmotic diarrhea. Two different ST enterotoxins have in humans is characterized by renal failure, widespread micro-
been identified, STa and STb. ST-producing ETEC have been thrombotic damage to a variety of organs, hemolytic anemia,
detected in young dogs with diarrhea.8-10 and sometimes diarrhea. Similar syndromes have been described
In addition to producing attaching and effacing lesions similar in dogs, but an associated E. coli infection was not demon-
to those created by EPEC, EHEC (Shiga-toxin–producing E. coli, strated.11,12 HUS has been reproduced experimentally in dogs
or STEC) produce Shiga-like toxins (also known as verotoxins). with a non-O157:H7 E. coli serotype.13 Based on the similarity
CHAPTER 46  Enteric Escherichia coli Infections 447

of renal lesions to those described in HUS, and the practice of


feeding predominantly raw meat diets to greyhounds, it has been
hypothesized that cutaneous and renal glomerular vasculopathy
of greyhounds (also known as “Alabama rot” or “Greenetrack
disease”) may result from infection with EHEC strains such as
O157:H7.14 A similar condition was described in a great Dane
from Germany.15 Some dogs develop cutaneous lesions in the
absence of renal failure; other dogs develop renal failure before
the onset of cutaneous lesions.
NTEC produce two toxins: CNF and cytolethal distending
toxin. Multiple CNF types have been identified (CNF1, CNF2,
and CNF3). NTEC have been associated with diarrhea, bacte-
remia, and urinary tract infections in human patients, as well as
with diarrhea in dogs.16
EIEC actively invade colonic epithelial cells. This, together
with the associated inflammatory response, leads to hemor-
rhagic, large bowel diarrhea. In human patients, the EIEC path-
ovar is uncommonly detected compared with EPEC and ETEC.
AIEC, which lack the virulence genes that identify EIEC, have FIGURE 46-2  Ulcerated and erythematous colon of a 2-year-old intact male boxer
been associated with granulomatous colitis in boxer dogs and dog with granulomatous colitis as viewed during colonoscopy. The dog had a 7-month
infrequently in the French bulldog and Border collie. This infec- history of marked weight loss and severe diarrhea, with increased frequency of defecation
tion typically affects young adult boxer dogs that show signs and hematochezia.
of severe colitis, with colonic thickening and ulceration, and
weight loss that is often marked.4,17 It has been suggested that a
heritable anomaly in boxer dogs predisposes them to the infec- microangiopathic hemolysis. Microcytic anemia may be present
tion, given the marked breed predisposition for the disease.18 in boxer dogs with granulomatous colitis.
The EAEC pathovar is considered an emerging pathogen
in human patients. It adheres to epithelial cells of the termi- Serum Chemistry Panel and Urinalysis
nal ileum and colon using fimbriae in a characteristic “stacked Laboratory abnormalities in dogs with E. coli diarrhea could
brick” pattern. Expression of these fimbriae is encoded by a include electrolyte abnormalities and prerenal azotemia in dogs
plasmid gene known as aggR. Bacterial aggregation is followed with severe diarrhea. Dogs infected with EHEC have had vari-
by damage and a subsequent inflammatory response, which able evidence of hypoalbuminemia, prerenal azotemia, increased
leads to development of persistent watery diarrhea. Occasion- activities of ALT and creatine kinase, hematuria, proteinuria,
ally, EAEC strains also produce toxins, such as verocytotoxin.19 and isosthenuria. Boxer dogs with granulomatous colitis are
Their role in dogs and cats requires further investigation. commonly hypoalbuminemic.

Physical Examination Findings Diagnostic Imaging


Physical examination may reveal dehydration and abdominal pain Findings on abdominal radiography and abdominal ultrasound
in puppies with E. coli diarrhea. Dogs with HUS have had cutane- are typically unremarkable or show fluid-filled intestinal loops.
ous erythema and well-demarcated, multifocal cutaneous ulcers of Abdominal ultrasound in boxer dogs with granulomatous coli-
the limbs. Fever and peripheral edema have also been described. tis often reveals mild or moderate mesenteric or sublumbar
Boxer dogs with granulomatous colitis may have poor body con- lymphadenomegaly, and the colonic wall can appear thickened.
dition. A thickened and irregular rectal wall may be detected on
rectal palpation, and the feces may contain fresh blood and mucus. Endoscopic Findings
Proctoscopy or colonoscopy is usually performed in order to
Diagnosis obtain colonic biopsies from boxer dogs suspected to have gran-
ulomatous colitis. Proctoscopy requires less fastidious colonic
Diagnosis of intestinal E. coli infections, with the exception of preparation compared to colonoscopy, can be performed under
granulomatous colitis of boxer dogs, is difficult or impossible heavy sedation, and is more cost effective compared to colo-
with currently available diagnostic assays, because of the fact noscopy. Most boxer dogs with granulomatous colitis have
that healthy dogs and cats shed E. coli, and it is a combination involvement of the descending colon, underscoring the diagnos-
of bacterial virulence factors and host immune competence that tic utility of proctoscopy. Common changes in appearance to
contributes to the development of clinical signs. This section the colonic wall include erythema, irregularity, and ulceration
therefore focuses primarily on the more specific clinical syn- of the rectal wall (Figure 46-2).
dromes caused by EHEC and AIEC.
Microbiologic Testing
Laboratory Abnormalities Bacterial Isolation and Typing
Complete Blood Count E. coli is commonly isolated on routine bacteriologic media
CBC changes in dogs with E. coli diarrhea are generally mild from feces of both healthy and diarrheic dogs. A differential
and nonspecific. Leukocytosis may be present. Dogs infected medium such as MacConkey agar is often used. Apart from
with EHEC may be thrombocytopenic and have evidence of dogs with granulomatous colitis, attempts to specifically iden-
tify E. coli as a cause of diarrhea are generally performed only
448 SECTION 2  Bacterial Diseases

by reference or public health laboratories when an outbreak has


occurred, or for research purposes. E. coli can be isolated from
colonic biopsies of dogs with granulomatous colitis, which
permits subsequent antimicrobial susceptibility testing.20 Sero-
logic identification, which is performed using specific O and
H antisera, can be costly and is generally performed by large
public health laboratories in epidemiologic investigations.

Virulence Assays
Immunoassays are available for detection of Shiga toxin, ST,
and LT. A Vero cell cytotoxicity assay has been used to detect
verocytotoxin. Identification of EPEC and AIEC often relies on
assays that assess adherence in tissue culture. In research set-
tings, molecular diagnostic techniques using DNA probes or
PCR assays have revolutionized the ability to detect and dif-
ferentiate between pathogenic and nonpathogenic strains of E.
coli. EPEC characteristically possess the eaeA gene. Detection of
genes that encode LT, ST, and Shiga-like toxin allows identifica- A
tion of ETEC and EHEC. EIEC are identified based on the pres-
ence of invasion-associated genes such as ipaH. The aat gene is
a diagnostic marker for EAEC detection.19

Pathologic Findings
The most significant pathologic findings in intestinal E. coli
infections occur with EHEC and AIEC infections.

EHEC
Cutaneous ulcerative lesions in dogs with suspected HUS are
characterized histopathologically by fibrinoid vascular necro-
sis, with dermal thrombosis and leukocytoclastic vasculitis.
Grossly, the kidneys may be swollen and have cortical pete-
chiae. Histopathology reveals hyaline fibrinous thrombi in
glomerular capillaries and afferent arterioles, glomerular and
multifocal tubular necrosis, and leukocytoclastic vasculitis with
fibrinoid necrosis. B

AIEC FIGURE 46-3  A, Histopathology showing severe, chronic, histiocytic colitis in a


2-year-old male boxer dog. The inflammatory infiltrate is comprised of large, epithelioid
Histopathologic lesions in boxers and French bulldogs with
macrophages and lesser numbers of neutrophils, plasma cells, and lymphocytes. B, Histio-
granulomatous colitis are pathognomonic and include neutro- cytes in the lamina propria contain PAS-positive material.
philic inflammation, epithelial ulceration, crypt hyperplasia and
distortion, decreased goblet cell numbers, and abundant macro-
phages that stain positive with periodic acid–Schiff (PAS) stain rifampin, or trimethoprim-sulfonamides, should be chosen for
(Figure 46-3). The presence of E. coli within macrophages can treatment based on the results of susceptibility testing.
be confirmed using fluorescence in situ hybridization, which has
been recommended as part of the diagnostic work-up of dogs Immunity and Vaccination
suspected to have histiocytic ulcerative colitis.5,18
Currently, no vaccines for E. coli infections are available for
Treatment and Prognosis dogs and cats. Vaccines for EPEC, ETEC, and EHEC have been
investigated for animals and humans on a research basis,21-23
Dogs and cats with severe diarrhea due to pathogenic E. coli including a transdermal LT vaccine for prevention of travelers’
may require intravenous fluid therapy. Dogs with granuloma- diarrhea.24
tous colitis can show dramatic responses to treatment with
enrofloxacin (10 mg/kg PO q24h), and a minimum treatment Prevention
duration of 8 weeks is recommended.18 Administration of fluo-
roquinolones is usually associated with rapid resolution of clini- The reader is referred to Chapter 11 and Chapter 45 for infor-
cal signs and resolves cellular infiltration characteristic of this mation on prevention of enteric bacterial infections.
disorder on colonic biopsy. Because enrofloxacin resistance has
been documented in some isolates from dogs with granuloma- Public Health Aspects
tous colitis, attempts to isolate E. coli from colonic biopsies
before treatment is recommended so that antimicrobial suscepti- Pathogenic E. coli that cause diarrhea have been particularly
bility testing can be performed.20 Antimicrobials that penetrate prevalent in children in developing countries and in travel-
intracellularly, such as fluoroquinolones, chloramphenicol, ers to developing countries. EHEC can be acquired following
CHAPTER 46  Enteric Escherichia coli Infections 449

ingestion of undercooked ground hamburger meat, vegetables, healthy companion animals can be shared with humans that
and nonchlorinated drinking and swimming water that have reside in the same household,29-32 but more studies are required
been contaminated with fecal material. Infection has also been to determine the extent to which this occurs and its zoonotic sig-
reported following contact with animals housed in petting nificance. Transfer of plasmids that carry resistance genes from
zoos.25 HUS is reported as the most common cause of acute canine or feline E. coli isolates and those of humans also has the
renal failure in children. Clinical signs in humans consist of potential to occur.
gastrointestinal signs, often with profuse, bloody diarrhea, For hospitalized animals, staff should use contact precau-
abdominal cramps, pallor, weakness, and oliguria or anuria.26 tions for any dog or cat suspected to have enteropathogenic
Dogs have been suggested as a source of human infection with illness, including the use of warning signs, gloves, and gowns,
EPEC and EHEC.27,28 hand washing, and cleaning with bleach-based or accelerated
The possibility of zoonotic transmission of multidrug resis- hydrogen peroxide-based disinfectants. Endoscopes should be
tant E. coli that are shed by dogs and cats is an emerging con- disinfected thoroughly between uses. Veterinarians should dis-
cern. A variety of resistance mechanisms have been identified in cuss the zoonotic implications of enteropathogenic bacterial
E. coli isolates from dogs and cats, which include production infection with pet owners, especially in relation to the presence
of efflux pumps and extended-spectrum and AmpC β-lactamase of young children or otherwise immunocompromised individu-
enzymes. There is some evidence that E. coli strains shed by als in the household.

CASE EXAMPLE
Signalment: “Caesar”, 2-year-old male boxer dog from
Tulare, CA (Figure 46-4)
History: Caesar was evaluated for chronic diarrhea, which
had been present since he was 6 months of age. His feces
were loose and contained a small amount of frank blood.
He strained to defecate many times a day, and the owner
sometimes noticed mucus in the diarrhea. Every few
months, Caesar’s appetite and energy level declined and
the diarrhea became more severe. His signs transiently
improved after treatment with subcutaneous fluids and
metronidazole. The owners had tried a variety of different
diets such as fiber-supplemented diets and elimination
diets that contain novel, single protein sources, but these
strategies did not help. His current diet was a digestible
commercial diet, and his appetite was good. He was up
to date on vaccinations, including those for rabies, canine
distemper, canine parvovirus, canine adenovirus, and
Bordetella bronchiseptica. He was primarily an indoor dog
with access to a suburban backyard.
Physical Examination:
Body Weight: 23.4 kg
General: Bright, alert, and responsive. Ambulatory on all four
limbs. T = 102.5°F (39.2°C), HR = 102 beats/min, panting, FIGURE 46-4  “Caesar,” a 2-year-old male boxer dog from Tulare, CA, that was
mucous membranes pink, CRT = 1 s. diagnosed with granulomatous colitis following colonoscopy and biopsy.
Integument, Eyes, Ears, Nose, and Throat: No significant
abnormalities were noted. A moderate amount of dental MCHC 32.9 g/dL (33-36 g/dL)
calculus was present. WBC 12,590 cells/µL (6000-13,000 cells/µL)
Musculoskeletal: Body condition score was 3/9 with Neutrophils 9631 cells/µL (3000-10,500 cells/µL)
symmetrical muscling. Lymphocytes 1674 cells/µL (1000-4000 cells/µL)
All Other Systems: On rectal examination, the walls of Monocytes 781 cells/µL (150-1200 cells/µL)
the rectum were smooth. There was fresh blood on the Platelets were clumped but appeared adequate in number.
glove following palpation. No other clinically significant Serum Chemistry Profile:
abnormalities were noted. Sodium 155 mmol/L (145-154 mmol/L)
Laboratory Findings: Potassium 4.2 mmol/L (3.6-5.3 mmol/L)
CBC: Chloride 116 mmol/L (108-118 mmol/L)
HCT 54.4% (40%-55%) Bicarbonate 23 mmol/L (16-26 mmol/L)
MCV 76.8 fL (65-75 fL) Phosphorus 4.4 mg/dL (3.0-6.2 mg/dL)
450 SECTION 2  Bacterial Diseases

Calcium 10.4 mg/dL (9.7-11.5 mg/dl) Histopathology of Colonic Biopsies: Severe, chronic,
BUN 18 mg/dL ( 5-21 mg/dL) histiocytic and ulcerative colitis. Histiocytes in the lamina
Creatinine 1.1 mg/dL (0.3-1.2 mg/dL) propria contained PAS-positive material (see Figure 46-3).
Glucose 105 mg/dL (64-123 mg/dL) Diagnosis: Granulomatous colitis of boxer dogs
Total protein 6.5 g/dL (5.4-7.6 g/dL) Treatment: Enrofloxacin, 6 mg/kg PO q24h for 8 weeks and
Albumin 2.9 g/dL (3.0-4.4 g/dL) a commercial low-residue dry and canned dog food. This
Globulin 3.6 g/dL (1.8-3.9 g/dL) was associated with clinical improvement within a week
ALT 68 U/L (19-67 U/L) after starting the medication. At a recheck 4 weeks later, the
AST 46 U/L (19-42 U/L) owner reported that Caesar’s feces were completely normal
ALP 86 U/L (21-170 U/L) in consistency and frequency.
GGT 5 U/L (0-6 U/L) Comments: In this dog, the diagnosis of granulomatous colitis
Cholesterol 200 mg/dL (135-361 mg/dL) was made solely based on histopathologic findings. Culture
Total bilirubin 0.1 mg/dL (0-0.2 mg/dL). of a biopsy specimen could also have been performed and is
Fecal Examination: Centrifugal fecal flotation: Negative for now recommended given the emergence of fluoroquinolone-
parasites. resistant AIEC strains of E. coli.
Colonoscopy: The entire mucosal surface of the colon was
erythematous and irregular, with multifocal areas of mucosal
ulceration (see Figure 45-1).

SUGGESTED READINGS 13. Wang JY, Wang SS, Yin PZ. Haemolytic-uraemic syndrome caused
by a non-O157:H7 Escherichia coli strain in experimentally inocu-
Sherman PM, Ossa JC, Wine E. Bacterial infections: new and emerging lated dogs. J Med Microbiol. 2006;55(Pt 1):23-29.
enteric pathogens. Curr Opin Gastroenterol. 2010;26(1):1-4. 14. Cowan LA, Hertzke DM, Fenwick BW, et al. Clinical and clinico-
Simpson KW, Dogan B, Rishniw M, et al. Adherent and invasive Esch- pathologic abnormalities in greyhounds with cutaneous and renal
erichia coli is associated with granulomatous colitis in boxer dogs. glomerular vasculopathy: 18 cases (1992-1994). J Am Vet Med
Infect Immun. 2006;74(8):4778-4792. Assoc 210(6):789–793.
15. Rotermund A, Peters M, Hewicker-Trautwein M, et  al. Cutane-
REFERENCES ous and renal glomerular vasculopathy in a great Dane resembling
1. Escherich T. Die Darmbakterien des Neugeborenen und Säuglings. “Alabama rot” of greyhounds. Vet Rec. 2002;151(17):510-512.
Fortschr Med. 1885;3:515-522, 547-554. 16. Starcic M, Johnson JR, Stell AL, et  al. Haemolytic Escherichia
2. Sancak AA, Rutgers HC, Hart CA, et al. Prevalence of enteropathic coli isolated from dogs with diarrhea have characteristics of
Escherichia coli in dogs with acute and chronic diarrhea. Vet Rec. both uropathogenic and necrotoxigenic strains. Vet Microbiol.
2004;154(4):101-106. 2002;85(4):361-377.
3. Morato EP, Leomil L, Beutin L, et  al. Domestic cats constitute 17. Van Kruiningen HJ, Civco IC, Cartun RW. The comparative
a natural reservoir of human enteropathogenic Escherichia coli importance of E. coli antigen in granulomatous colitis of Boxer
types. Zoonoses Public Health. 2009;56(5):229-237. dogs. APMIS. 2005;113(6):420-425.
4. Simpson KW, Dogan B, Rishniw M, et al. Adherent and invasive 18. Mansfield CS, James FE, Craven M, et al. Remission of histiocytic
Escherichia coli is associated with granulomatous colitis in boxer ulcerative colitis in Boxer dogs correlates with eradication of invasive
dogs. Infect Immun. 2006;74(8):4778-4792. intramucosal Escherichia coli. J Vet Intern Med. 2009;23(5):964-969.
5. Celli J, Deng W, Finlay BB. Enteropathogenic Escherichia coli 19. Scavia G, Staffolani M, Fisichella S, et al. Enteroaggregative Esch-
(EPEC) attachment to epithelial cells: exploiting the host cell cyto- erichia coli associated with a foodborne outbreak of gastroenteritis.
skeleton from the outside. Cell Microbiol. 2000;2(1):1-9. J Med Microbiol. 2008;57(Pt 9):1141-1146.
6. Morato EP, Leomil L, Beutin L, et  al. Domestic cats constitute 20. Craven M, Dogan B, Schukken A, et  al. Antimicrobial resistance
a natural reservoir of human enteropathogenic Escherichia coli impacts clinical outcome of granulomatous colitis in boxer dogs.
types. Zoonoses Public Health. 2009;56:229-237. J Vet Intern Med. 2010;24(4):819-824.
7. de Almeida PM, Arais LR, Andrade JR, et al. Characterization of 21. Gu J, Liu Y, Yu S, et  al. Enterohemorrhagic Escherichia coli tri-
atypical Enteropathogenic Escherichia coli (aEPEC) isolated from valent recombinant vaccine containing EspA, intimin and Stx2
dogs. Vet Microbiol. 2012;158:420-424. induces strong humoral immune response and confers protection in
8. Drolet R, Fairbrother JM, Harel J, et al. Attaching and effacing and mice. Microbes Infect. 2009;11(10-11):835-841.
enterotoxigenic Escherichia coli associated with enteric colibacil- 22. Keller R, Hilton TD, Rios H, et  al. Development of a live oral
losis in the dog. Can J Vet Res. 1994;58(2):87-92. attaching and effacing Escherichia coli vaccine candidate using
9. Hammermueller J, Kruth S, Prescott J, et  al. Detection of toxin Vibrio cholerae CVD 103-HgR as antigen vector. Microb Pathog.
genes in Escherichia coli isolated from normal dogs and dogs with 2010;48(1):1-8.
diarrhea. Can J Vet Res. 1995;59(4):265-270. 23. Rojas RL, Gomes PA, Bentancor LV, et  al. Salmonella enterica
10. Beutin L. Escherichia coli as a pathogen in dogs and cats. Vet Res. serovar Typhimurium vaccine strains expressing a nontoxic Shiga-
1999;30(2-3):285-298. like toxin 2 derivative induce partial protective immunity to the
11. Chantrey J, Chapman PS, Patterson-Kan JC. Haemolytic-­uraemic toxin expressed by enterohemorrhagic Escherichia coli. Clin Vac-
syndrome in a dog. J Vet Med A Physiol Pathol Clin Med. cine Immunol. 2010;17(4):529-536.
2002;49(9):470-472. 24. Frech SA, Dupont HL, Bourgeois AL, et al. Use of a patch contain-
12. Dell’Orco M, Bertazzolo W, Pagliaro L, et  al. Hemolytic-uremic ing heat-labile toxin from Escherichia coli against traveller’s diar-
syndrome in a dog. Vet Clin Pathol. 2005;34(3):264-269. rhoea: a phase II, randomized, double-blind, placebo-controlled
field trial. Lancet. 2008;371(9629):2019-2025.
CHAPTER 46  Enteric Escherichia coli Infections 451

25. Sherman PM, Ossa JC, Wine E. Bacterial infections: new and emerg- 30. Stenske KA, Bemis DA, Gillespie BE, et al. Comparison of clonal
ing enteric pathogens. Curr Opin Gastroenterol. 2010;26(1):1-4. relatedness and antimicrobial susceptibility of Escherichia coli from
26. Scheiring J, Andreoli SP, Zimmerhackl LB. Treatment and outcome healthy dogs and their owners. Am J Vet Res. 2009;70:1108-1116.
of Shiga-toxin-associated hemolytic-uremic syndrome. Pediatr 31. Johnson JR, Clabots C, Kuskowski MA. Multiple-host sharing,
Nephrol. 2008;23(10):1749-1760. long-term persistence, and virulence of Escherichia coli clones
27. Nakazato G, Gyles C, Ziebell K, et al. Attaching and effacing Esch- from human and animal household members. J Clin Microbiol.
erichia coli isolated from dogs in Brazil: characteristics and sero- 2008;46:4078-4082.
typic relationship to human enteropathogenic E. coli (EPEC). Vet 32. Damborg P, Nielsen SS, Guardabassi L. Escherichia coli shedding
Microbiol. 2004;101(4):269-277. patterns in humans and dogs: insights into within-household trans-
28. Hogg RA, Holmes JP, Ghebrehewet S, et  al. Probable zoonotic mission of phylotypes associated with urinary tract infections. Epi-
transmission of verocytotoxigenic Escherichia coli O157 by dogs. demiol Infect. 2012;137:1457-1464.
Vet Rec. 2009;164(10):304-305.
29. Harada K, Okada E, Shimuzu T, et  al. Antimicrobial resistance,
virulence profiles and phylogenetic groups of fecal Escherichia coli
isolates: a comparative analysis between dogs and their owners in
Japan. Comp Immunol Microbiol Infect Dis. 2012;35:139-144.
CHAPTER 47

Campylobacteriosis
Jane E. Sykes and Stanley L. Marks

healthy and diarrheic dogs and cats. In one study, Campylo-


Overview of Campylobacteriosis bacter species were isolated more commonly from healthy cats
First Described: Germany, 1886 (Theodor Escherich)1; the than from cats with diarrhea.3
organism was not isolated until the 1950s C. jejuni, C. helveticus, and C. upsaliensis are the species
most frequently isolated from the stool of dogs and cats, with
Cause: Campylobacter spp. (curved to spiral gram-negative
C. helveticus and C. upsaliensis being the predominant isolates
bacteria that belong to the Campylobactereaceae)
in some studies.3,4 Other commonly identified species in dogs
Affected Hosts: Humans and a large variety of other animal and cats include Campylobacter coli, C. helveticus, and Campy-
species lobacter lari. In one study, up to 12 Campylobacter species were
Geographic Distribution: Worldwide present simultaneously in dogs,5 and coinfection with multiple
Campylobacter species was detected in 47% of 74 cats from
Primary Mode of Transmission: Fecal-oral
which Campylobacter was isolated.3 The prevalence of Campy-
Major Clinical Signs: Fever, lethargy, anorexia, large bowel lobacter carriage is particularly high in young dogs and cats and
diarrhea those housed with large numbers of other animals such as in
Differential Diagnoses: Differential diagnoses for suspected kennels or shelters. Increased isolation rates are also observed in
Campylobacter enterocolitis include canine and feline par- the spring, summer, and fall months, depending on study loca-
vovirus infection, canine distemper virus infection, salmon tion.6,7 Cats less than 1 year of age were significantly more likely
­poisoning disease, Escherichia coli infection, clostridial to shed Campylobacter than cats older than a year of age, and
diarrhea, salmonellosis, giardiasis, tritrichomoniasis (cats), median duration of shedding was around 6 weeks in a study from
cryptosporidiosis, whipworms, leptospirosis, dietary indis- the upper Midwestern United States.7 Young age and the feeding
cretion, gastrointestinal foreign body, pancreatitis, inflam- of homemade cooked food to dogs was associated with Campy-
matory bowel disease, lymphoma, ­hyperthyroidism (cats), lobacter carriage in dogs from eastern Canada.2 Ownership of
hypoadrenocorticism, toxins (including drugs). puppies and kittens is a significant risk factor for development
of campylobacteriosis by humans, especially young children.8,9
Human Health Significance: Important zoonosis. Dogs and cats
may be a source of human infection with Campylobacter, Clinical Features
some of which may be resistant to antimicrobial drugs.
Signs and Their Pathogenesis
Campylobacter colonize the lower intestinal tract, including the
Etiology and Epidemiology jejunum, ileum, and colon. In contrast to other enteropathogenic
bacteria, Campylobacter possess relatively few virulence factors,
Campylobacter are thin, gram-negative, curved, S-shaped or and host factors are important in determining the severity of
spiral rods that are often motile by means of a single polar clinical signs that develop. Campylobacter can invade intestinal
flagellum at one or both ends (Figure 47-1). They are micro- epithelial cells and produce cytolethal distending toxin, which
aerophilic (i.e., they grow best when levels of oxygen are lower causes cell cycle arrest and apoptosis, and in humans, stimulates
than that present in the atmosphere), and are widespread in the Il-8 production, which leads to an inflammatory response. The
gastrointestinal tracts of animals. Campylobacter spp. have the organism can produce a polysaccharide capsule, which may also
potential to be zoonotic and are an important cause of food- be a virulence factor.10 Puppies and kittens less than 6 months
borne illness in humans, especially in association with poultry of age are most likely to show diarrhea in association with Cam-
products. They can survive weeks to months in water. More pylobacter infection, and stress, overcrowding, and concurrent
than a dozen species have been identified in dogs and cats (Box gastrointestinal infections with other bacteria, protozoa, and hel-
47-1). All species are commonly isolated from both healthy and minth parasites may also contribute to clinical signs. The sever-
diarrheic dogs and cats. The prevalence of shedding by clinically ity of diarrhea can range from mild diarrhea with loose stools
healthy dogs, for example, ranges from 15% to 87%,2 and as through to watery, bloody, or mucoid diarrhea, with lethargy,
high as 58% of non-diarrheic and diarrheic cats shed Campy- inappetence, and, less commonly, vomiting. Other signs of large
lobacter as determined by PCR assay.3 Campylobacter jejuni is bowel diarrhea such as tenesmus may also be present. Fever may
most commonly incriminated as a cause of diarrhea. The evi- occur in severe acute campylobacteriosis in puppies and kittens.
dence for disease causation by other species, such as Campylo- Diarrhea is generally self-limiting within a 1- to 2-week period,
bacter upsaliensis and Campylobacter helveticus, is less certain, although chronic diarrhea can also occur in dogs.11,12 Rarely,
because the isolation rates of these species are often similar in extraintestinal Campylobacter infections in dogs have been

452
CHAPTER 47  Campylobacteriosis 453

ATL
0
1
GB 2
FF 3
4

5
6

FIGURE 47-1  Scanning electron micrograph of Campylobacter. (From Centers for Dis-
FIGURE 47-2  Abdominal ultrasound image from a 10-year-old intact male ­Brittany
ease Control and Prevention, Atlanta, GA.)
spaniel with Campylobacter bacteremia, gastroenteritis, and probable cholecystitis, ­showing
thickening and irregularity of the gallbladder (GB) wall and free abdominal fluid (FF).

BOX 47-1 Diagnostic Imaging


Findings on plain abdominal radiography and abdominal ultra-
Campylobacter Species Described in Dogs and Cats sound are typically unremarkable or show fluid-filled intestinal
loops. A thickened gallbladder wall may be seen using ultrasound
C. jejuni in animals with Campylobacter cholecystitis (Figure 47-2).
C. upsaliensis
C. lari Microbiologic Testing
C. coli Microbiologic assays available for diagnosis of campylobacteri-
C. helveticus osis in dogs and cats include microscopic examination, culture,
C. concisus and PCR assays (Table 47-1). Because of the high prevalence of
C. fetus Campylobacter colonization in healthy dogs and cats, it is dif-
C. gracilis ficult to interpret the significance of positive test results on feces.
C. mucosalis The results must always be considered in light of the patient
C. showae signalment, history, clinical signs, and exclusion of other causes
C. sputorum of diarrhea. Specific identification of C. jejuni, as opposed to
C. curvus other Campylobacter species, may also add support to the role
C. hyointestinalis of Campylobacter in disease.
C. rectus
Microscopic Examination
Although not diagnostic for campylobacteriosis, a direct fecal
smear may reveal large numbers of fine, S-shaped or gull-shaped
reported, specifically cholecystitis, bacteremia, and abortion.13,14 organisms following staining with Gram or Romanowsky stains
These have also been described in humans.15,16 (Figure 47-3). Detection of these organisms only suggests the
presence of Campylobacter-like organisms and should not be
Physical Examination Findings used as the sole method to diagnose campylobacteriosis because
Physical examination findings in severely affected young ani- of the inability to differentiate between similar-appearing
mals with campylobacteriosis are variable and include lethargy, organisms such as Arcobacter or nonpathogenic campylobac-
dehydration, fever, and abdominal pain. Diarrhea, sometimes ters. Fecal leukocytes may also be present. More advanced
with fresh blood or mucus, may be found on rectal examination microscopic techniques that have been used to identify Cam-
in dogs. pylobacter in the feces include darkfield and phase-contrast
microscopy, which are used on fresh fecal specimens and show
Diagnosis the characteristic morphology and darting motility of the organ-
ism. These are generally not performed routinely in clinical situ-
Laboratory Abnormalities ations and require significant technical expertise.
Complete Blood Count and Chemistry Panel
Basic laboratory testing in puppies and kittens with Campylo- Bacterial Isolation
bacter diarrhea generally reveals mild and nonspecific changes. Campylobacter cannot be isolated on routine bacteriological
A leukocytosis may be present. Animals with systemic cam- media. Fecal enteric panels designed to detect bacterial entero-
pylobacteriosis and cholecystitis may show neutrophilia or pathogens in feces generally include isolation on selective
neutropenia with increased circulating band neutrophils and media such as charcoal- or blood-based Campylobacter media,
biochemical evidence of cholestasis and hepatic dysfunction (see which often contain antimicrobials to eliminate other bacteria
Case Example). (such as cefoperazone). The inoculated medium is incubated
454 SECTION 2  Bacterial Diseases

TABLE 47-1
Diagnostic Assays Available for Campylobacteriosis in Dogs and Cats
Assay Specimen Type Target Performance
Bacterial isolation Feces, blood, bile, Campylobacter False negatives may occur, especially following antimicrobial therapy,
intestinal tissues organism and some species may not grow under routine conditions used to
collected at isolate Campylobacter. Isolation from feces does not i­mply that
n­ecropsy Campylobacter is the cause of disease. Allows subsequent antimi-
crobial susceptibility testing.
Polymerase chain As for isolation Campylobacter Rapid (within hours); can be highly sensitive and specific. Positive
reaction DNA results from feces do not imply that Campylobacter is the cause of
disease. Some assays do not differentiate between Campylobacter
species, but others are species specific (e.g., only Campylobacter
jejuni or only Campylobacter coli). Check with the laboratory to
determine what assays are offered.

rapid identification of Campylobacter species and strains (see


Chapter 3).21,22 Isolation of Campylobacter in culture permits
subsequent antimicrobial susceptibility testing, although this is
not routinely performed by most veterinary laboratories.

Molecular Diagnosis Using the Polymerase Chain Reaction


Both conventional and real-time PCR-based assays have been
developed for direct detection of Campylobacter in stool, and
some are available commercially for use in dogs and cats. An
advantage of PCR is that it can detect species that grow poorly
in culture, and it overcomes problems relating to overgrowth
of other fecal bacteria.3 Both Campylobacter genus-specific and
species-specific assays (such as those that only detect C. jejuni)
have been developed. In human patients, a real-time PCR assay
for detection of C. jejuni was at least as sensitive as culture.23

Pathologic Findings
Gross Pathologic Findings
Gross necropsy findings in puppies with campylobacteriosis
include fluid-filled intestinal loops with mucosal congestion and
edema, especially within the distal jejunum, ileum, cecum, and
FIGURE 47-3  Cytology showing Campylobacter organisms. Gram stain, 1000× colon.12,24 Mesenteric lymphadenomegaly may also be present.
magnification.
Histopathologic findings
in a microaerophilic environment for 72 to 96 hours. Incuba- Campylobacter infection can result in villous blunting and
tion is frequently carried out at 42°C to select for thermophilic fusion, epithelial hyperplasia, and congestion and lymphoplas-
­Campylobacter; however, a temperature of 37°C should also be macytic inflammation of the lamina propria. Crypts contain
used to ensure isolation of variable or nonthermophilic species. filamentous bacteria that stain with silver stains such as War-
The use of direct plating onto charcoal-based media may be most thin-Starry (Figure 47-4). Immunohistochemistry can be used to
sensitive for isolation of Campylobacter from pets, but use of a identify Campylobacter within intestinal crypts.12
combination of isolation methods increases sensitivity further.17
Some strains require more hydrogen than others and do not Treatment and Prognosis
grow under routine conditions used for Campylobacter isola-
tion. Species identification can then be performed on the basis Uncomplicated campylobacteriosis is generally self-limiting and
of biochemical testing or the use of PCR followed by sequencing resolves with supportive therapy. Because isolation of Campylo-
of the PCR product. Biochemical testing can be highly variable, bacter even from diarrheic feces does not necessarily imply causa-
resulting in inaccurate identification. Serotyping has been tradi- tion for the clinical signs, specific treatment may not be warranted
tionally used to identify strains of C. jejuni for epidemiologic and may further disrupt the intestinal microflora. Treatment could
studies, and more recently, molecular methods such as pulsed- be considered for severely ill dogs and cats. Macrolides (such as
field gel electrophoresis and PCR-based typing methods (includ- erythromycin or azithromycin) and fluoroquinolones (such as
ing multilocus sequence typing), have been used.18-20 Techniques enrofloxacin, 5 mg/kg q24h for 5 to 7 days) are often efficacious,
based on matrix-assisted laser desorption/ionization-time-of- although resistance to these antimicrobials has been documented
flight (MALDI-TOF) mass spectrometry also show promise for in some Campylobacter isolates.25,26 Treatment failures could
CHAPTER 47  Campylobacteriosis 455

reflect resistance to the antimicrobial used or infection with a Drinking water and swimming water may also be a source of
nonpathogenic Campylobacter species and persistence of clinical infection. To a lesser extent, contact with other food animal spe-
signs from another unidentified cause. Recrudescence of infection cies, wild birds, and pet dogs and cats has been associated with
has been documented in human patients after treatment.27 human campylobacteriosis.32 Ingestion of as few as 500 organ-
isms can lead to infection. When clinical signs occur, early signs
Immunity and Vaccination develop after an incubation period of 1 to 7 days and include
fever, myalgia, vomiting, and headache, which generally last
Innate, humoral, and cell-mediated immune responses are 1 to 3 days, followed by up to one week of watery to bloody diar-
critical for clearance of Campylobacter infection.28 Short-term rhea and abdominal pain. Illness may be mild to severe, requiring
protection that is somewhat serotype specific can result from hospitalization. Severe illness is more likely to occur in the immu-
frequent exposure to Campylobacter, but immunity wanes with nosuppressed. Reactive arthritis and Guillain-Barré syndrome,
time, and reinfection is possible. Considerable effort has been an acute inflammatory demyelinating polyneuropathy, are late-
expended into development of vaccines for humans and poul- onset immune-mediated consequences that can sometimes occur
try.29-31 No vaccine currently exists for prevention of Campylo- in association with infection and may persist for weeks or years.32
bacter infection in dogs and cats. In fact, C. jejuni is the most common infection that precedes Guil-
lain-Barré syndrome, which occurs in 1 in 1000 infected individu-
Public Health Aspects als.25 Extraintestinal infections that have been described include
bacteremia, hepatitis, cholecystitis, pancreatitis, peritonitis, abor-
Campylobacter is the most common bacterial cause of enteric tion and neonatal sepsis, urinary tract infections, myocarditis,
illness in humans worldwide. Over 90% of human campylo- and meningitis, although these are very rare in immunocompe-
bacteriosis in industrialized countries results from consumption tent individuals. Campylobacter enteritis has also been identified
of contaminated chicken products, as well as beef and milk. as a risk factor for inflammatory bowel disease.33

A B C
FIGURE 47-4  Histopathology of an intestinal section from a 3-month-old Chihuahua puppy with Campylobacter jejuni–associated diarrhea. Abundant filamentous, spiral bacteria
pack the crypts and there is an associated lymphoplasmacytic enterocolitis. A, H&E stain; B, Giemsa stain; C, Warthin-Starry silver stain.

CASE EXAMPLE Integument, Eyes, Ears, Nose, and Throat: A large amount of
dental calculus was noted. There was moderate waxy debris
Signalment: “Henry,” a 10-year-old intact male Brittany in both ear canals and pinnal erythema.
spaniel from Dixon in northern California Musculoskeletal: Body condition score was 4/9, and the dog
History: Henry was brought to an emergency clinic for a 1-day was ambulatory with symmetrical muscling.
history of anorexia, lethargy, and fever. The owner, a retired Cardiovascular: The femoral pulses were fair and synchronous.
veterinarian, noted that Henry’s rectal temperature was 106°F No murmurs or arrhythmias were auscultated.
(41.1°C). There had been no vomiting or diarrhea, and the dog Respiratory: Harsh breath sounds were bilaterally noted on
had been drinking normally. Henry was a field trial dog and thoracic auscultation, which were loudest in the ventral lung
was hunting a week before becoming ill. He was housed in fields.
an outdoor run with no access to foreign objects other than Gastrointestinal and Genitourinary: The abdomen was
gravel. There were nine other apparently healthy dogs in the tense on palpation and moderate hepatomegaly was noted.
household. The dogs were fed a commercial dry and canned The urinary bladder was large. No abnormalities were
diet. detected on rectal examination.
Physical Examination: Lymph Nodes: All were within normal limits.
Body Weight: 17.8 kg Laboratory Findings:
General: Quiet, alert and responsive, 5% to 7% dehydrated. CBC:
Ambulatory. T = 106.4°F (41.3°C), HR = 140 beats/min, RR = HCT 42.9% (40%-55%)
40 breaths/min, mucous membranes pale pink and tacky, MCV 70 fL (65-75 fL)
CRT = 2 s. MCHC 34.5 g/dL (33-36 g/dL)
456 SECTION 2  Bacterial Diseases

The stomach and duodenal wall were mildly thickened.


Moderate mesenteric lymphadenopathy was identified,
particularly around the liver and ileocolic junction. The
prostate was enlarged with multiple small parenchymal
cysts, but was symmetric and smoothly margined. A sample
of orange, somewhat turbid abdominal fluid and an aspirate
of a mesenteric lymph node were obtained using ultrasound
guidance.
Cytology Findings: Abdominal fluid: Modified transudate.
TP = 4.8 g/dL, 20,000 RBC/µL, 460 nucleated cells/µL, with
72% neutrophils, 21% small mononuclear cells, and 7%
large mononuclear cells. The nucleated cells were primarily
nondegenerate neutrophils with smaller numbers of
lymphocytes and macrophages. The lymphocytes were a
heterogenous population of small mature lymphocytes
and scattered granular lymphocytes. Several pyknotic cells
and rare erythrophagocytic macrophages were present. No
FIGURE 47-5  Abdominal radiograph from a 10-year-old intact male Brittany
spaniel with Campylobacter bacteremia, gastroenteritis, and probable cholecystitis infectious organisms were seen.
that shows hepatomegaly and corrugation of the colonic wall. Mesenteric lymph node: Mild lymphoid reactivity.
Microbiologic Testing: Vector-borne disease serologic
2 nucleated red blood cells/100 WBC testing (IFA): Positive for serum Babesia canis antibodies
WBC 6600 cells/µL (6,000-13,000 cells/µL) (1:160), negative for antibodies to Rickettsia rickettsii,
Neutrophils 2904 cells/µL (3000-10,500 cells/µL) Anaplasma phagocytophilum, and Ehrlichia canis.
Band neutrophils 2838 cells/µL Aerobic and anaerobic bacterial culture of abdominal fluid: No
Metamyelocytes 330 cells/µL growth.
Lymphocytes 330 cells/µL (1000-4000 cells/µL) Aerobic bacterial urine culture (cystocentesis specimen): No
Monocytes 198 cells/µL (150-1200 cells/µL) growth.
Eosinophils 0 cells/µL (0-1500 cells/µL) Aerobic and anaerobic blood culture (four specimens collected
Platelets 144,000/µL (150,000-400,000 platelets/µL). over 5 hours, the first inoculated into a blood culture bottle
Neutrophils, band neutrophils, and metamyelocytes and the remainder into isolator tubes): Direct smear of blood
showed mild toxic changes. culture bottle contents showed rare small curved gram-neg-
Serum Chemistry Profile: ative rods. Anaerobic culture of first specimen showed small
Sodium 146 mmol/L (145-154 mmol/L) curved gram-negative rods suspicious for Campylobacter
Potassium 4.1 mmol/L (3.6-5.3 mmol/L) spp. Partial sequencing of the 16S rRNA gene revealed 100%
Chloride 116 mmol/L (108-118 mmol/L) identity to Campylobacter jejuni. All other specimens yielded
Bicarbonate 15 mmol/L (16-26 mmol/L) no growth.
Phosphorus 3.9 mg/dL (3.0-6.2 mg/dL) Fecal enteric panel: Clostridium difficile TcdA and TcdB toxin ELISA
Calcium 8.6 mg/dL (9.7-11.5 mg/dl) negative. ELISA negative for Clostridium perfringens entero-
BUN 21 mg/dL ( 5-21 mg/dL) toxin. No Salmonella spp. cultured. No Clostridium difficile cul-
Creatinine 0.8 mg/dL (0.3-1.2 mg/dL) tured. Small numbers of Campylobacter spp. cultured (C. jejuni
Glucose 86 mg/dL (64-123 mg/dL) using partial 16S rRNA gene sequencing).
Total protein 5.0 g/dL (5.4-7.6 g/dL) Diagnosis: Campylobacter jejuni bacteremia and probable
Albumin 2.7 g/dL (3.0-4.4 g/dL) cholangiohepatitis/cholecystitis
Globulin 2.3 g/dL (1.8-3.9 g/dL) Treatment and Outcome: Henry was hospitalized and
ALT 794 U/L (19-67 U/L) treated aggressively with intravenous fluids, and before
AST 275 U/L (19-42 U/L) culture results became available, enrofloxacin (5 mg/kg
ALP 461 U/L (21-170 U/L) IV q12h) and ampicillin (22 mg/kg IV q8h). Shortly after
GGT 37 U/L (0-6 U/L) admission he vomited and developed mucus-containing
Cholesterol 273 mg/dL (135-361 mg/dL) diarrhea, and subsequently melena was observed. Over
Total bilirubin 0.7 mg/dL (0-0.2 mg/dL). the next 48 hours his temperature gradually normalized,
Urinalysis: SGr 1.013 (after fluid bolus), pH 7.5, 2+ protein but he continued to vomit intermittently. Neutropenia
(SSA), 1+ bilirubin, 1+ hemoprotein, no glucose, rare WBC/ persisted, and his platelet count dropped to 105,000. He
HPF, rare RBC/HPF, rare granular casts. also became icteric, and there were progressive increases
Imaging Findings: in the activities of liver enzymes (ALT 1179 U/L, AST 946
Thoracic and Abdominal Radiographs: The cardiovascular U/L, ALP 1088 U/L, GGT 15 U/L, total bilirubin 3.9 g/dL). By
and pulmonary structures were within normal limits. The day 4 after presentation, vomiting and diarrhea ceased and
liver was moderately enlarged. The colon was mildly gas he began eating a bland diet. Laboratory testing showed
distended and had a corrugated appearance (Figure 47-5). resolution of neutropenia and thrombocytopenia. He was
Abdominal Ultrasound: There was moderate enlargement of discharged from the hospital and treated with enrofloxacin
the liver and spleen. The gallbladder wall was thickened and (10 mg/kg PO q24h) for 4 weeks. One week after discharge,
mildly irregular. There was moderate peritoneal effusion. Henry was doing well and had a normal appetite and
CHAPTER 47  Campylobacteriosis 457

activity level. The owner reported that another dog in the was not performed, but might also have been helpful. Had
household subsequently developed vomiting and diarrhea, C. jejuni only been isolated from the dog’s feces, its clinical
and Campylobacter spp. were cultured from the dog’s feces. significance would have been less apparent. Whether or not
Comments: This was an unusual case of C. jejuni bacteremia the other dog in the household truly had campylobacteriosis
and probable cholangiohepatitis/cholecystitis in a dog that is unknown.
required blood culture for definitive diagnosis. Culture of bile

SUGGESTED READINGS 16. Udayakumar D, Sanaullah M. Campylobacter cholecystitis. Int J


Med Sci. 2009;6(6):374-375.
Butzler JP. Campylobacter, from obscurity to celebrity. Clin Microbiol 17. Acke E, McGill K, Golden O, et al. A comparison of different cul-
Infect. 2004;10(10):868-876. ture methods for the recovery of Campylobacter species from pets.
Chaban B, Ngeleka M, Hill JE. Detection and quantification of 14 Cam- Zoonoses Public Health. 2009;56(9-10):490-495.
pylobacter species in pet dogs reveals an increase in species richness in 18. Parsons BN, Cody AJ, Porter CJ, et al. Typing of Campylobacter jejuni
feces of diarrheic animals. BMC Microbiol. 2010;10:73. isolates from dogs by use of multilocus sequence typing and pulsed-
Dasti JI, Tareen AM, Lugert R, et  al. Campylobacter jejuni: a brief field gel electrophoresis. J Clin Microbiol. 2009;47(11):3466-3471.
overview on pathogenicity-associated factors and disease-mediating 19. Cornelius AJ, Gilpin B, Carter P, et al. Comparison of PCR binary
mechanisms. Int J Med Microbiol. 2010;300(4):205-211. typing (P-BIT), a new approach to epidemiological subtyping of
Campylobacter jejuni, with serotyping, pulsed-field gel electro-
REFERENCES phoresis, and multilocus sequence typing methods. Appl Environ
1. Escherich T. Über das Vorkommen von Vibrionen im Darmkanal Microbiol. 2010;76(5):1533-1544.
und den Stuhlgängen der Säuglinge. Münch Med Wochenschr. 20. Parsons BN, Porter CJ, Stavisky JH, et al. Multilocus sequence typ-
1886;33:815-817, 833-835. ing of human and canine Campylobacter upsaliensis isolates. Vet
2. Leonard EK, Pearl DL, Janecko N, et  al. Factors related to Microbiol. 2012;157:391-397.
Campylobacter spp. carriage in client-owned dogs visiting vet- 21. Kiehntopf M, Melcher F, Hänel I, et al. Differentiation of Campylo-
erinary clinics in a region of Ontario, Canada. Epidemiol Infect. bacter species by surface-enhanced laser desorption/ionization-time-
2011;139:1531-1541. of-flight mass spectrometry. Foodborne Pathog Dis. 2011;8:875-885.
3. Queen EV, Marks SL, Farver TB. Prevalence of selected bacterial 22. Bessède E, Solecki O, Sifré E, et al. Identification of Campylobacter
and parasitic agents in feces from diarrheic and healthy control cats species and related organisms by matrix-assisted laser desorption/
from northern California. J Vet Intern Med. 2012;26(1):54-60. ionization-time-of-flight (MALDI-TOF) mass spectrometry. Clin
4. Parsons BN, Porter CJ, Ryvar R, et  al. Prevalence of Campylo- Microbiol Infect. 2011;17:1735-1739.
bacter spp. in a cross-sectional study of dogs attending veterinary 23. Lin S, Wang X, Zheng H, et al. Direct detection of Campylobacter
practices in the UK and risk indicators associated with shedding. jejuni in human stool samples by real-time PCR. Can J Microbiol.
Vet J. 2010;184(1):66-70. 2008;54(9):742-747.
5. Chaban B, Ngeleka M, Hill JE. Detection and quantification of 14 24. Fox JG, Maxwell KO, Ackerman JI. Campylobacter jejuni asso-
Campylobacter species in pet dogs reveals an increase in species ciated diarrhea in commercially reared beagles. Lab Anim Sci.
richness in feces of diarrheic animals. BMC Microbiol. 2010;10:73. 1984;34(2):151-155.
6. Sandberg M, Bergsjo B, Hofshagen M, et al. Risk factors for Cam- 25. Butzler JP. Campylobacter, from obscurity to celebrity. Clin Micro-
pylobacter infection in Norwegian cats and dogs. Prev Vet Med. biol Infect. 2004;10(10):868-876.
2002;55:241-253. 26. Lindow JC, Poly F, Tribble DR, et  al. Caught in the act: in  vivo
7. Bender JB, Shulman SA, Averbeck GA, et  al. Epidemiologic fea- development of macrolide resistance to Campylobacter jejuni infec-
tures of Campylobacter infection among cats from the western tion. J Clin Microbiol. 2010;48(8):3012-3015.
United States. J Am Vet Med Assoc. 2005;226(4):544-547. 27. Baqar S, Tribble DR, Carmolli M, et  al. Recrudescent Campylo-
8. Wolfs TF, Dulm B, Geelen SP, et  al. Neonatal sepsis by Campy- bacter jejuni infection in an immunocompetent adult following
lobacter jejuni: genetically proven transmission from a household experimental infection with a well-characterized organism. Clin
puppy. Clin Infect Dis. 2001;32(5):E97-E99. Vaccine Immunol. 2010;17(1):80-86.
9. Deming MS, Tauxe RV, Blake PA, et al. Campylobacter enteritis at 28. Janssen R, Krogfelt KA, Cawthraw SA, et al. Host-pathogen inter-
a university: transmission from eating chicken and from cats. Am J actions in Campylobacter infections: the host perspective. Clin
Epidemiol. 1987;126(3):526-534. Microbiol Rev. 2008;21(3):505-518.
10. Guerry P, Poly F, Riddle M, et al. Campyobacter polysaccharide cap- 29. Jagusztyn-Krynicka EK, Łaniewski P, Wyszyńska A. Update on
sules: virulence and vaccines. Front Cell Infect Microbiol. 2012;2:7. Campylobacter jejuni vaccine development for preventing human
11. Davies AP, Gebhart CJ, Meric SA. Campylobacter-associated chronic campylobacteriosis. Expert Rev Vaccines. 2009;8(5):625-645.
diarrhea in a dog. J Am Vet Med Assoc. 1984;184(4):469-471. 30. Tribble DR, Baqar S, Carmolli MP, et  al. Campylobacter jejuni
12. Brown C, Martin V, Chitwood S. An outbreak of enterocolitis strain CG8421: a refined model for the study of campylobacteriosis
due to Campylobacter spp. in a beagle colony. J Vet Diagn Invest. and evaluation of Campylobacter vaccines in human subjects. Clin
1999;11(4):374-376. Infect Dis. 2009;49(10):1512-1519.
13. Odendaal MW, de Cramer KG, van der Walt ML, et al. First isola- 31. Buckley AM, Wang J, Hudson DL, et  al. Evaluation of live-­
tion of Campylobacter jejuni from the vaginal discharge of three attenuated Salmonella vaccines expressing Campylobacter antigens for
bitches after abortion in South Africa. Onderstepoort J Vet Res. control of C. jejuni in poultry. Vaccine. 2010;28(4):1094-1105.
1994;61(2):193-195. 32. Dasti JI, Tareen AM, Lugert R, et al. Campylobacter jejuni: a brief
14. Bulgin MS, Ward AC, Sriranganathan N, et al. Abortion in the dog overview on pathogenicity-associated factors and disease-mediating
due to Campylobacter species. Am J Vet Res. 1984;45(3):555-556. mechanisms. Int J Med Microbiol. 2010;300(4):205-211.
15. Smith JL. Campylobacter jejuni infection during pregnancy: long- 33. Kalischuk LD, Buret AG. A role for Campylobacter jejuni–induced
term consequences of associated bacteremia, Guillain-Barré syn- enteritis in inflammatory bowel disease? Am J Physiol Gastrointest
drome, and reactive arthritis. J Food Prot. 2002;65(4):696-708. Liver Physiol. 2010;298(1):G1-G9.
CHAPTER 48

Enteric Clostridial Infections


Jane E. Sykes and Stanley L. Marks

veterinary hospitals.4,5 In one study, living with an immuno-


Overview of Clostridial Diarrhea compromised human was a risk factor for C. difficile infection
First Described: 1978 (C. difficile, causative role in pseudo- in dogs, but strains present in dogs differed from those in the
membranous colitis in humans)1; 1892 (C. perfringens, by household environment.6 Dogs that visit human health care
Nuttall and Welch).2 Evidence for a role of C. perfringens in facilities are also at risk of acquiring C. difficile infections.7 The
food-borne illness was gathered over 50 years later.3 emergence of “hypervirulent” C. difficile strains in people has
Cause: Clostridium difficile and Clostridium perfringens (gram- resulted in an increased incidence of disease, increased mortality
positive, spore-forming anaerobic rods) rates, increased relapse rates, and recognition of community-­
associated disease.8 A similar change has not been observed
Affected Hosts: Humans and a variety of other animals
in animals; however, a hypervirulent strain (North American
Geographic Distribution: Worldwide pulsotype 1 or NAP1) was identified in a dog.9 C. difficile
Primary Mode of Transmission: Fecal-oral; disease can result strains have been classified on the basis of PCR-based typing
from toxin production by organisms resident in the gas- of their ribosomal RNA genes (PCR-ribotyping). In one study,
trointestinal tract multiple animals in shelter environments were colonized with
a single PCR-ribotype (SLO 066, 045, and 010 in three differ-
Major Clinical Signs: Lethargy, anorexia, diarrhea, occasion-
ent ­shelters, respectively).10 More detailed genetic analysis of
ally fever
the C. difficile strains isolated by multilocus variable number
Differential Diagnoses: Differential diagnosis for suspected of tandem repeat analysis (MLVA) suggested that horizontal
clostridial enterocolitis include canine and feline parvo- transmission of strains 045 and 010 occurred within the shelter.
virus infection, canine distemper virus infection, salmon C. perfringens is especially widespread in soil and dust and
poisoning disease, campylobacteriosis, salmonellosis, consists of five biotypes, A to E, based on the possession of
giardiasis, tritrichomoniasis (cats), cryptosporidiosis, whip- one or more of four major toxin genes (encoding alpha, beta,
worms, leptospirosis, dietary indiscretion, gastrointestinal ­epsilon, and iota toxins). Each biotype can also express a subset
foreign body, pancreatitis, inflammatory bowel disease, of at least 10 other established toxins, including C. perfringens
lymphoma, hypoadrenocorticism, toxins (including drugs) enterotoxin (CPE). Almost all infections in dogs and cats are
Human Health Significance: Dogs and cats have the potential C. perfringens biotype A, which also causes the majority of clos-
to be a source of human infection with C. difficile, because tridial food-borne illness in humans. The zoonotic potential of
they can harbor strains (ribotypes) that have been C. difficile and C. perfringens is still incompletely understood.
detected in humans. The role of dogs and cats as a source
of C. perfringens infection is poorly understood.

Etiology and Epidemiology


Clostridium difficile and Clostridium perfringens are gram-­
positive, spore-forming, obligately anaerobic rods that are
capable of toxin production. Clostridial spores are spherical
to oval and distend the bacterial cells when they form within
them (­Figure 48-1). Clostridium species are found in soil and the
intestinal tracts of a variety of different animal species, including
those of healthy dogs and cats. Their spores are very resistant to
disinfection and can persist in the environment, including within
hospitals, for years. Despite their similarities, many differences
exist between C. difficile and C. perfringens that relate to epide-
miology, interpretation of diagnostic test results, and treatment.
C. difficile is the most common cause of hospital and
antimicrobial-associated diarrhea in humans and has also FIGURE 48-1  Fecal smear from a dog with diarrhea showing spore-distended
been associated with nosocomial diarrhea in dogs and cats in ­clostridial rods, leading to a characteristic “safety pin” and “tennis racquet” appearance.

458
CHAPTER 48  Enteric Clostridial Infections 459

The importance of C. perfringens and C. difficile as causes


of diarrhea in dogs and cats has been controversial, because
they can be isolated from both diarrheic and healthy animals.
For example, C. perfringens can be isolated using culture from
the feces of more than 70% of both diarrheic and nondiarrheic
dogs.11,12 The prevalence of C. perfringens colonization of
healthy cats appears to be lower than that in dogs, with isola-
tion rates of 43% to 63%.13 Similarly, toxin production can be
detected in both healthy and diarrheic dogs and cats, although
most studies have shown a correlation between production of
certain clostridial toxins and diarrhea.11

Clinical Features
Signs and Their Pathogenesis
Both C. difficile and C. perfringens cause diarrhea by produc- FIGURE 48-2  Abdominal ultrasound image showing thickening and corrugation of
ing toxins. The diarrhea can be mild and self-limiting or severe, the small intestinal wall of a 2-year-old intact male chihuahua with Clostridium perfrin-
acute, and hemorrhagic, with life-threatening consequences that gens–associated vomiting, hematemesis, diarrhea, and hematochezia. Large numbers of
spore-forming gram-positive rods were seen on a fecal smear. C. perfringens enterotoxin
relate to dehydration and hypovolemic shock. Inappetence,
was detected in the feces using an ELISA assay.
vomiting, fever, and abdominal pain have also been described
in cats with C. difficile-associated diarrhea.14 Vomiting can also
occur in dogs. Although clostridial diarrhea is often described The prevalence of C. perfringens–associated diarrhea in cats is
as a large bowel–type diarrhea, clinical manifestations of both much lower than in dogs, and CPE has been detected at a simi-
small and large bowel diarrhea have been well documented. lar rate in diarrheic and nondiarrheic cats (2% to 4.1%).13 In
C. difficile can produce up to five toxins, the most well studied another study, CPE was detected with ELISA in 19.6% of 46
being toxin A (TcdA), toxin B (TcdB), and C. difficile binary toxin diarrheic dogs versus 2.1% of 48 healthy dogs, and in 8.3%
(CDT, an ADP-ribosyltransferase that is made up of two compo- of 48 diarrheic cats versus 0% of 39 healthy cats.21 In a PCR-
nents). CDT-positive strains have not been reported in dogs and based study of cats and dogs in an animal shelter in Florida,
cats. In contrast to humans, a history of antibiotic use does not the C. perfringens alpha toxin gene was detected in 64% of 50
always seem to be necessary for colonization and disease in dogs diarrheic dogs and 40% of 50 nondiarrheic dogs, which was
and cats, although in one study administration of antimicrobials significantly different.22 In contrast, it was detected in 42% of
before admission and the use of immunosuppressive drugs during 50 diarrheic and 50% of 50 nondiarrheic cats in the same shel-
hospitalization were risk factors for nosocomial colonization.4 ter.23 Thus the role that C. perfringens plays in feline diarrhea,
Toxins A and B are thought to be most important in the patho- if any, remains unclear.
genesis of diarrhea.15 These are large toxins that inactivate the
Rho family of GTPases, which regulate intracellular actin dynam- Physical Examination Findings
ics. The result is a disruption of the cell cytoskeleton, with subse- Physical examination findings in dogs and cats with severe clos-
quent rounding and death of intestinal epithelial cells, and loss of tridial diarrhea include dehydration, fever, and abdominal pain,
tight junctions. The two toxins also cause release of inflammatory with or without signs of hypovolemic shock. Rectal examination
cytokines from mast cells, macrophages, and epithelial cells.15 may reveal loose or watery stools, sometimes with fresh blood
In dogs, C. perfringens infection has been associated with and mucus.
nosocomial diarrhea,16 hemorrhagic enteritis,17-19 and acute
and chronic large bowel diarrhea. C. perfringens diarrhea in Diagnosis
dogs and cats is thought to occur subsequent to massive prolif-
eration and sporulation of resident clostridial organisms in the Laboratory Abnormalities
intestinal lumen following an alteration in local intestinal condi- Complete Blood Count and Chemistry Panel
tions. Decreased peristalsis, the effects of antimicrobial drugs on Basic laboratory testing in dogs and cats with clostridial diarrhea
the resident intestinal microflora, diet changes, and/or co-infec- usually reveals mild and nonspecific changes. A neutrophilic leu-
tions with other intestinal pathogens has the potential to trigger kocytosis with a left shift and toxic changes may be present in
sporulation. C. perfringens produces CPE on sporulation, but dogs and cats with acute clostridial gastroenteritis. In addition, a
other toxins and bacterial virulence factors may also be involved discordant hematocrit and serum total protein concentration can
in the pathogenesis of diarrhea, because diarrhea can occur in be identified in some dogs with C. perfringens and C. difficile-
the absence of CPE production. CPE is a pore-forming protein associated diarrhea. The hematocrit can be markedly increased
toxin that is released when vegetative clostridial cells lyse and in the face of a normal or low-normal total protein.
release their spores. It binds primarily to claudin, a component
of epithelial tight junctions, which leads to increased paracellu- Diagnostic Imaging
lar permeability.20 The role of CPE in the development of diar- Findings on plain abdominal radiography and abdominal
rhea in dogs and cats is unclear, because CPE is detected in 34% ultrasound examination in animals with clostridial diarrhea
of diarrheic dogs, and in 5% to 14% of nondiarrheic dogs.11,12 are typically unremarkable or show fluid-filled intestinal
There may be an association between the detection of CPE in loops. Mild thickening and corrugation of the small intestinal
dogs with acute hemorrhagic diarrheal syndrome (AHDS), or colonic wall may be detected on ultrasound examination
as CPE was detected in 8 of 12 dogs (67%) with AHDS.18 (Figure 48-2).
460 SECTION 2  Bacterial Diseases

TABLE 48-1
Diagnostic Assays Available for Clostridial Enterocolitis in Dogs and Cats
Specimen
Assay Type Target Performance
Bacterial Feces C. perfringens and C. difficile Isolation from feces does not imply that Clostridium is the cause of
­isolation disease. Allows genotyping and subsequent antimicrobial suscep-
tibility testing if needed. False negatives may occur, especially
for C. difficile, which requires strict anaerobic conditions.
Common Feces C. difficile Excellent negative predictive value for C. difficile (i.e., a negative
­antigen test test result suggests C. difficile is not present). A positive test
result does not imply that C. difficile is the cause of disease.
Toxin immu- Feces C. perfringens enterotoxin Positive test results may occur in dogs and cats that do not have
noassays (CPE); C. difficile toxin A diarrhea; negative results do not rule out clostridial diarrhea, as
and toxin B other toxins may be involved and CPE is relatively labile.
PCR Feces Clostridium toxin gene DNA Rapid (within hours). Positive results from feces do not imply
that Clostridium is the cause of disease. Toxin gene PCR assays
may be useful for diagnosis in conjunction with positive ELISA
results for toxin production. Negative results may occur if other
toxin genes are important or if inhibitors of PCR are present.

Microbiologic Testing Median spore counts of greater than 106 per gram of feces col-
Diagnostic assays available for clostridial enterocolitis in dogs lected within 48 hours after the onset of illness are suggestive
and cats are shown in Table 48-1. Because organisms and their of infection.25 Such counts can still be detected in humans with
toxins can be found in healthy and diarrheic animals, the results no signs of illness, and the method, when used without assess-
must be interpreted in light of the history, clinical signs, and ment of toxin production, has not proven useful for diagnosis of
diagnostic testing to rule out other causes of disease. C. perfringens diarrhea in dogs.12

Microscopic Examination ELISA for C. difficile Antigen


Spore-forming rods may be identified on microscopic examina- C. difficile antigen ELISA is easy to perform, rapid, and highly
tion of fecal smears that have been stained with Wright stains sensitive. The antigen test detects “common antigen” (glutamate
(see Figure 48-1). Rods that contain endospores have the appear- dehydrogenase; GDH) that is predominantly present in toxi-
ance of “tennis racquets” or “safety pins.” However, there is no genic and nontoxigenic C. difficile strains and a few uncommon
association between fecal endospore counts and the presence Clostridium spp. This test has the same limitations as culture in
of diarrhea, or between endospore counts and the detection of terms of detection of nontoxigenic strains, although a negative
CPE in fecal specimens. Detection of fecal endospores is thus an test for GDH can virtually rule out infection with C. difficile.
unreliable test for diagnosis of C. perfringens diarrhea in dogs
and cats and should not be used as a stand-alone test to make Assays for Toxin Production
clinical inferences in diarrheic dogs and cats.11,24 Several commercial serologic assays are available for detec-
Bacterial isolation.  Both C. difficile and C. perfringens can tion of C. difficile toxin A and toxin B in feces. Fecal swab
be isolated from feces of dogs and cats using strict anaerobic specimens generally do not contain a sufficient quantity of
conditions for incubation. C. perfringens is more tolerant of stool for these assays. Even when performed on larger vol-
oxygen than C. difficile and therefore may be easier to isolate umes of stool, the sensitivities and specificities of these assays
(hence the name difficile, because of its difficulty to isolate in vary considerably, and the etiologic predictive values are rela-
the laboratory). tively low for all tests. In other words, a positive result does
C. difficile can be isolated from 0% to 40% of diarrheic and not prove that a clostridial infection is the cause of a dog or
healthy dogs. When fecal culture for C. difficile is performed cat’s diarrhea. Currently, assays based on ELISA methodology
on a viable fecal specimen by a reputable laboratory, a nega- have shown the best correlation in dogs between clostridial
tive result suggests that C. difficile is not present. However, a toxin detection and the presence of diarrhea in dogs.11,16,24
positive culture does not differentiate between toxigenic and Panels should include assays for both C. difficile toxin A and
nontoxigenic strains, and further testing for toxin is warranted. toxin B, because TcdA-negative, TcdB-positive strains have
Because of the high carriage rate of C. perfringens in both been documented in dogs, and production of both toxins is
healthy and diarrheic dogs (>70% in both groups), routine not needed for disease causation.26 The reference standard in
isolation of C. perfringens is not recommended for diagnosis, human medicine for C. difficile toxin production is a cell cul-
unless epidemiologic studies that require genetic typing are ture cytotoxicity assay, which uses Vero cells and neutralizing
undertaken in outbreak situations. Quantitative stool spore antibody to inhibit cytopathic effects.27,28 However, culture
counts have been used to aid diagnosis of C. perfringens diar- followed by molecular detection of toxin genes within the iso-
rhea in humans. The stool is treated with heat or alcohol after lated strains (also known as “toxigenic culture”) appears to
which it is cultured anaerobically and colonies are counted. be a more sensitive reference standard.28 Both of these assays
CHAPTER 48  Enteric Clostridial Infections 461

are time consuming and expensive, require specific techni- Treatment and Prognosis
cal expertise, and are of limited availability. The use of mul-
tiple assays in sequence has been advocated for diagnosis of Animals with suspected clostridial enterocolitis that are sys-
C. difficile infection in humans.29 Specifically, fecal isolation temically ill (fever, hemorrhagic gastroenteritis, inflammatory
of C. difficile or detection of common antigen should be fol- or toxic leukogram) merit appropriate antimicrobial therapy
lowed with ELISA testing for toxin A and toxin B. Detection (Table 48-2). There is no documented evidence for a benefit of
of toxin but failure to identify C. difficile should be inter- antimicrobial therapy in dogs with uncomplicated Clostridium-
preted with caution, considering the relatively high sensitivity associated diarrhea, and most of these animals can be managed
of organism or common antigen detection methods compared supportively to ensure adequate hydration status without anti-
to toxin ELISA. microbial therapy. In fact, a prospective, blinded study that ran-
Detection of C. perfringens enterotoxin in stool is the most domized dogs with hemorrhagic gastroenteritis that showed no
widely used diagnostic tool for C. perfringens infection in both signs of sepsis to two groups, one treated with clavulanic acid–
humans and animals. Two commercially available immunoas- amoxicillin for 7 days and one treated with placebo, showed no
says are currently used in veterinary diagnostic laboratories: a difference in mortality rate, dropout rate, duration of hospital-
reverse passive latex agglutination assay (RPLA) and an ELISA. ization, or severity of clinical signs, either on any individual day
The assays have not been validated in animals to date. In addi- or over the course of disease.32
tion, the RPLA has been associated with false-positive results Metronidazole is the drug of choice for treatment of com-
when compared to several different ELISA methods. plicated C. difficile infections. Oral vancomycin (which is not
absorbed from the gastrointestinal tract) is occasionally used in
Molecular Diagnosis Using the Polymerase Chain Reaction human medicine to treat metronidazole-resistant isolates. The
Real-time PCR assays are commercially available for detection vast majority of C. perfringens isolates from dogs are sensitive
of C. perfringens and C. difficile toxin genes in the feces of dogs to metronidazole, ampicillin, amoxicillin, tylosin, and macrolides
and cats. It is important to recognize that the presence of clos- such as erythromycin.33 Because of the infrequency of resistance
tridial toxin genes within fecal specimens does not prove that to these drugs, the lack of known breakpoints for veterinary
clostridia are the cause of an animal’s diarrhea, because they do anaerobes, and the lack of relationship between positive stool cul-
not prove that the organism is actively producing toxin within tures and disease, antimicrobial susceptibilities are not performed
the intestinal tract of a patient. Conversely, a negative PCR result on a routine basis for these organisms when isolated from dogs
for clostridial toxin genes does not rule out clostridial diarrhea, and cats. Most animals with complicated clostridial enterocolitis
as clostridia may be present that possess other virulence factors, can be successfully treated with antimicrobials for 5 to 7 days,
and inhibitors of PCR are common in fecal specimens. Real- and appropriate treatment is generally associated with clinical
time PCR assays for C. difficile toxin genes have been devel- improvement within 24 to 36 hours.
oped for diagnosis of C. difficile diarrhea in humans.30 Some
assays can detect mutations within the toxin gene that correlate Immunity and Vaccination
with increased toxin production. However, it is still not well
understood how well the results of these assays correlate with No vaccines are available to prevent clostridial diarrhea in dogs
clinical illness. Similarly, detection of the cpe gene has been used and cats.
to diagnose C. perfringens diarrhea in humans.31 In one study,
fecal specimens from diarrheic dogs were more likely to be posi- Public Health Aspects
tive for CPE as detected using ELISA and PCR for the cpe gene
(28%) when compared with nondiarrheic dogs (4%). Such a C. difficile is increasingly recognized as an important cause
combination of testing for the toxin (CPE) via ELISA and the of nosocomial diarrhea and pseudomembranous colitis in
enterotoxin gene (cpe) via PCR is likely superior to either test humans, especially the elderly, which is induced by antimicro-
alone.12 At the time of writing, real-time PCR assays that pro- bial treatment (especially ampicillin, amoxicillin, clindamycin,
vide quantitative information on toxin gene copy numbers are cephalosporins, and fluoroquinolones)34 or disruption of the
available through some veterinary diagnostic laboratories. The normal gastrointestinal flora. Humans with C. difficile infec-
degree to which toxin gene copy numbers correlate with clinical tion develop bloating, and often foul-smelling diarrhea with
signs requires further study. abdominal pain, usually 5 to 10 days after starting antibiotics.

TABLE 48-2
Antimicrobials That Can Be Used to Treat Complicated Clostridial Diarrhea in Dogs and Cats*
Drug Dose (mg/kg) Route Interval (hours) Duration (days)
Metronidazole 10 PO 12 5-7
Ampicillin 22 PO 8-12 5-7
Amoxicillin 22 PO 12 5-7
Tylosin 5 to 10 PO 24 5-7
Erythromycin 15 PO 8 5-7

*Antimicrobial treatment of uncomplicated clostridial diarrhea is not recommended.


462 SECTION 2  Bacterial Diseases

Other signs include fever, nausea, anorexia, and malaise. Rare meat and companion animal food products in transmission of
and life-threatening complications of pseudomembranous the organism.6,37
colitis include acute toxic megacolon, an acute dilatation of C. perfringens infections are associated with sporadic and anti-
the colon with signs of obstruction, and colonic perforation. microbial-associated diarrhea in humans and CPE is one of the most
Hypervirulent strains (such as PCR-ribotype 027) that produce commonly identified causes of food poisoning in the industrialized
CDT, toxin A, and toxin B have been increasingly identified world.38 Clinical signs of infection include foul-smelling, frothy
recently that cause a more severe disease, higher relapse rates, diarrhea, cramping and abdominal pain, and occasionally nausea
and increased mortality.35 Efforts are underway to develop vac- and vomiting, usually 7 to 15 hours after eating suspected food,
cines for C. difficile infections in humans.36 There is evidence especially meat products that have been cooked with improper
that dogs may shed C. difficile strains similar to those that infect cooling and reheating.39 Vegetative cells are then ingested, and this
humans. Human colonization and disease usually occurs in hos- can be followed by massive sporulation with toxin production in
pital situations, and community-acquired infection is uncom- the intestinal tract. The zoonotic potential of C. perfringens iso-
mon, although concerns have been raised regarding the role of lates from dogs and cats requires further investigation.

CASE EXAMPLE Laboratory Findings:


CBC:
HCT 46% (40%-55%)
Signalment: “Bradley”, 2-year old male Chihuahua from MCV 68 fL (65-75 fL)
Grass Valley in northern California MCHC 36.3 g/dL (33-36 g/dL)
History: Bradley was brought to an emergency clinic for WBC 11,030 cells/µL (6000-13,000 cells/µL)
evaluation of a 12-hour history of hematemesis and Neutrophils 7280 cells/µL (3000-10,500 cells/µL)
diarrhea. He initially vomited up dog food, after which he Band neutrophils 1324 cells/µL
continued to vomit hourly, and the vomiting progressed to Lymphocytes 1655 cells/µL (1000-4000 cells/µL)
hematemesis. He also developed bloody diarrhea. He had Monocytes 772 cells/µL (150-1200 cells/µL)
been inappetent and not drinking, and the owner felt he was Eosinophils 0 cells/µL (0-1500 cells/µL)
much quieter than usual. He primarily resided indoors and in Platelets 273,000/µL (150,000-400,000 platelets/µL).
the owner’s backyard, and there was no history of travel or Neutrophils and band neutrophils showed mild to moderate
access to toxins. There were two other apparently healthy toxic changes.
dogs in the household. He was up to date on vaccinations Serum Chemistry Profile:
for canine parvovirus, distemper virus, canine adenovirus, Sodium 147 mmol/L (145-154 mmol/L)
and rabies. His diet consisted of commercial dry dog food, Potassium 4.5 mmol/L (3.6-5.3 mmol/L)
dog bones, and table scraps. Chloride 111 mmol/L (108-118 mmol/L)
Physical Examination: Bicarbonate 29 mmol/L (16-26 mmol/L)
Body Weight: 3.4 kg Phosphorus 3.9 mg/dL (3.0-6.2 mg/dL)
General: Quiet, alert and responsive, ambulatory, 5% to 7% Calcium 9.6 mg/dL (9.7-11.5 mg/dl)
dehydrated. Recumbent. T = 100.0°F (37.8°C), HR = 100 BUN 15 mg/dL (5-21 mg/dL)
beats/min, RR = 30 breaths/min, mucous membranes pink Creatinine 0.4 mg/dL (0.3-1.2 mg/dL)
and tacky, CRT = 2 s. Glucose 123 mg/dL (64-123 mg/dL)
Integument: A full coat with a small amount of flea excrement Total protein 5.1 g/dL (5.4-7.6 g/dL)
was present. Bloody diarrhea contaminated the hair in the Albumin 3.1 g/dL (3.0-4.4 g/dL)
perianal region. Globulin 2.0 g/dL (1.8-3.9 g/dL)
Eyes, Ears, Nose, and Throat: No abnormalities were noted. ALT 68 U/L (19-67 U/L)
Musculoskeletal: Body condition score was 4/9. AST 31 U/L (19-42 U/L)
Cardiovascular: The femoral pulses were fair and synchronous. ALP 30 U/L (21-170 U/L)
No murmurs or arrhythmias were auscultated. GGT < 3 U/L (0-6 U/L)
Respiratory: Normal breath sounds were present in all lung Cholesterol 168 mg/dL (135-361 mg/dL)
fields. Total bilirubin 0.1 mg/dL (0-0.2 mg/dL).
Gastrointestinal and Genitourinary: The dog was Urinalysis: SGr 1.042, pH 6.0, 1+ protein (SSA), 1+ bilirubin,
nonpainful on abdominal palpation, but defecated red, no hemoprotein, no glucose, 1-2 WBC/HPF, 0 RBC/HPF, rare
jelly-like material several times in the room following the granular casts.
examination. He also urinated a large amount of odiferous Coagulation Panel: PT 8.9 s (7.5-10.5 s), APTT 18.1 s (9-12 s);
urine during the examination. No abnormalities were fibrinogen 349 mg/dL (90-255 mg/dL).
detected on rectal examination apart from the abnormal Imaging Findings: Abdominal ultrasound: Mild mesenteric
stool color and consistency. and sublumbar lymphadenopathy was identified. The
Lymph Nodes: All lymph nodes were within normal limits on small intestines had mild wall thickening and a corrugated
palpation. appearance (see Figure 48-2). The colon was fluid filled.
CHAPTER 48  Enteric Clostridial Infections 463

Fecal Examination: Fecal flotation: Negative for parasites. (7.5 mg/kg IV q12h). Vomiting did not continue, but red
Fecal sedimentation: Negative for parasites. gelatinous diarrhea continued every hour for the next
Microbiologic Testing: Parvovirus fecal antigen ELISA: 12 hours. Within 24 hours, he had become bright and alert
Negative. and was offered a bland diet, which he ate well. He was
Fecal direct smear: Large numbers of gram positive rods were discharged and treated with metronidazole for an additional
present, some with spores. 5 days, with gradual reintroduction of his regular food.
Fecal enteric panel: Clostridium difficile toxin A and toxin B anti- Comments: Infection by C. perfringens was strongly suspected
gen ELISA negative. ELISA positive for Clostridium perfringens in this case based on the combination of consistent
enterotoxin. No Salmonella spp. cultured. No Clostridium dif- clinical signs, negative assay results for other pathogens, a
ficile cultured. No Campylobacter spp. cultured. positive toxin ELISA assay, and response to treatment with
Diagnosis: Suspected C. perfringens–associated gastroenteritis. metronidazole. However, because the feces of healthy dogs
Treatment: Bradley was initially hospitalized and treated with can contain C. perfringens enterotoxin, a definitive diagnosis
intravenous fluids, nothing by mouth, and metronidazole of C. perfringens enterocolitis could not be made.

SUGGESTED READINGS 13. Marks SL, unpublished observations, 2011.


14. Weese JS, Weese HE, Bourdeau TL, et  al. Suspected Clostridium
Marks SL, Kather EJ, Kass PH, et  al. Genotypic and phenotypic difficile–associated diarrhea in two cats. J Am Vet Med Assoc.
­characterization of Clostridium perfringens and Clostridium difficile 2001;218(9):1436-1439,1421.
in diarrheic and healthy dogs. J Vet Intern Med. 2002;16(5):533-540. 15. Carter GP, Rood JI, Lyras D. The role of toxin A and toxin B in
Weese JS, Armstrong J. Outbreak of Clostridium difficile–associated Clostridium difficile–associated disease: past and present perspec-
disease in a small animal veterinary teaching hospital. J Vet Intern tives. Gut Microbes. 2010;1(1):58-64.
Med. 2003;17(6):813-816. 16. Kruth SA, Prescott JF, Welch MK, et al. Nosocomial diarrhea asso-
Weese JS, Staempfli HR, Prescott JF, et al. The roles of Clostridium dif- ciated with enterotoxigenic Clostridium perfringens infection in
ficile and enterotoxigenic Clostridium perfringens in diarrhea in dogs. dogs. J Am Vet Med Assoc. 1989;195(3):331-334.
J Vet Intern Med. 2001;15(4):374-378. 17. Sasaki J, Goryo M, Asahina M, et al. Hemorrhagic enteritis associ-
ated with Clostridium perfringens type A in a dog. J Vet Med Sci.
REFERENCES 1999;61(2):175-177.
1. George RH, Symonds JM, Dimock F. Identification of C. difficile as 18. Cave NJ, Marks SL, Kass PH, et al. Evaluation of a routine diag-
a cause of pseudomembranous colitis. Br Med J. 1978;i:695. nostic fecal enteric panel for dogs with diarrhea. J Am Vet Med
2. Welch WH, Nuttall GHF. A gas-producing bacillus (Bacillus aero- Assoc. 2002;221:52-59.
genes capsulatus, nov. spec.) capable of rapid development in blood 19. Schlegel BJ, Van Dreumel T, Slavić D, et al. Clostridium ­perfringens
vessels after death. Bull Johns Hopkins Hosp. 1892;3:81-91. type A fatal acute hemorrhagic gastroenteritis in a dog. Can Vet J.
3. Hobbs BC, Smith ME, Oakley CL, et al. Clostridium welchii food 2012;53:555-557.
poisoning. J Hyg. 1953;51(1):75-101. 20. McClane BA. The complex interactions between Clostridium
4. Weese JS, Armstrong J. Outbreak of Clostridium difficile–­ perfringens enterotoxin and epithelial tight junctions. Toxicon.
associated disease in a small animal veterinary teaching hospital. J 2001;39(11):1781-1791.
Vet Intern Med. 2003;17(6):813-816. 21. Leutenegger CM, Marks SL, Robertson J. Toxin quantification of
5. Clooten J, Kruth S, Arroyo L, et al. Prevalence and risk factors for Clostridium perfringens is a predictor for diarrhea in dogs and cats
Clostridium difficile colonization in dogs and cats hospitalized in [Abstract]. J Vet Intern Med. 2012;26:794.
an intensive care unit. Vet Microbiol. 2008;129(1-2):209-214. 22. Tupler T, Levy JK, Sabshin SJ, et al. Enteropathogens identified in
6. Weese JS, Finley R, Reid-Smith RJ, et al. Evaluation of Clostridium dogs entering a Florida animal shelter with normal feces or diar-
difficile in dogs and the household environment. Epidemiol Infect. rhea. J Am Vet Med Assoc. 2012;241:338-343.
2010;138(8):1100-1104. 23. Sabshin SJ, Levy JK, Tupler R, et al. Enteropathogens identified in
7. Lefebvre SL, Reid-Smith RJ, Waltner-Toews D, et al. Incidence of cats entering a Florida animal shelter with normal feces or diarrhea.
acquisition of methicillin-resistant Staphylococcus aureus, Clos- J Am Vet Med Assoc. 2012;241:331-337.
tridium difficile, and other health-care-associated pathogens by 24. Marks SL, Melli A, Kass PH, et al. Evaluation of methods to diag-
dogs that participate in animal-assisted interventions. J Am Vet nose Clostridium perfringens–associated diarrhea in dogs. J Am
Med Assoc. 2009;234(11):1404-1417. Vet Med Assoc. 1999;214(3):357-360.
8. Pepin J, Alary M, Valiquette L, et al. Increasing risk of relapse after 25. Birkhead G, Vogt RL, Heun EM, et al. Characterization of an out-
treatment of Clostridium difficile colitis in Quebec, Canada. Clin break of Clostridium perfringens food poisoning by quantitative
Infect Dis. 2005;40:1591-1597. fecal culture and fecal enterotoxin measurement. J Clin Microbiol.
9. Lefebvre S, Arroyo L, Weese J. Epidemic Clostridium difficile strain 1988;26(3):471-474.
in hospital visitation dog. Emerging Infect Dis. 2006;12:1036-1037. 26. Kuehne SA, Cartman ST, Heap JT, et  al. The role of toxin
10. Schneedberg A, Rupnik M, Neubauer H, et al. Prevalence and distri- A and toxin B in Clostridium difficile infection. Nature.
bution of Clostridium difficile PCR ribotypes in cats and dogs from 2010;467(7316):711-713.
animal shelters in Thuringia, Germany. Anaerobe. 2012;18:484-488. 27. Meer RR, Songer JG, Park DL. Human disease associated with
11. Weese JS, Staempfli HR, Prescott JF, et al. The roles of Clostridium Clostridium perfringens enterotoxin. Rev Environ Contam Toxi-
difficile and enterotoxigenic Clostridium perfringens in diarrhea in col. 1997;150:75-94.
dogs. J Vet Intern Med. 2001;15(4):374-378. 28. Crobach MJ, Dekkers OM, Wilcox MH, et  al. European Society
12. Marks SL, Kather EJ, Kass PH, et  al. Genotypic and pheno- of Clinical Microbiology and Infectious Diseases (ESCMID): data
typic characterization of Clostridium perfringens and Clos- review and recommendations for diagnosing Clostridium difficile-
tridium difficile in diarrheic and healthy dogs. J Vet Intern Med. infection (CDI). Clin Microbiol Infect. 2009;15(12):1053-1066.
2002;16(5):533-540.
464 SECTION 2  Bacterial Diseases

29. Schmidt ML, Gilligan PH. Clostridium difficile testing algorithms: 35. Cartman ST, Heap JT, Kuehne SA, et  al. The emergence of
what is practical and feasible? Anaerobe. 2009;15(6):270-273. “hypervirulence” in Clostridium difficile. Int J Med Microbiol.
30. Sloan LM, Duresko BJ, Gustafson DR, et al. Comparison of real- 2010;300(6):387-395.
time PCR for detection of the tcdC gene with four toxin immuno- 36. Lee BY, Popovich MJ, Tian Y, et al. The potential value of Clos-
assays and culture in diagnosis of Clostridium difficile infection. tridium difficile vaccine: an economic computer simulation model.
J Clin Microbiol. 2008;46(6):1996-2001. Vaccine. 2010;28(32):5245-5253.
31. Loh JP, Liu YC, Chew SW, et al. The rapid identification of Clos- 37. Gould LH, Limbago B. Clostridium difficile in food and domes-
tridium perfringens as the possible aetiology of a diarrhoeal out- tic animals: a new foodborne pathogen? Clin Infect Dis.
break using PCR. Epidemiol Infect. 2008;136(8):1142-1146. 2010;51(5):577-582.
32. Unterer S, Strohmeyer K, Kruse BD, et  al. Treatment of aseptic 38. Brynestad S, Granum PE. Clostridium perfringens and food-borne
dogs with hemorrhagic gastroenteritis with amoxicillin/clavulanic infections. Int J Food Microbiol. 2002;74(3):195-202.
acid: a prospective blinded study. J Vet Intern Med. 2011;25(5): 39. Shandera WX, Tacket CO, Blake PA. Food poisoning due
973-979. to Clostridium perfringens in the United States. J Infect Dis.
33. Marks SL, Kather EJ. Antimicrobial susceptibilities of canine Clos- 1983;147(1):167-170.
tridium difficile and Clostridium perfringens isolates to commonly
utilized antimicrobial drugs. Vet Microbiol. 2003;94(1):39-45.
34. Pépin J, Saheb N, Coulombe MA, et  al. Emergence of fluoroqui-
nolones as the predominant risk factor for Clostridium difficile–
associated diarrhea: a cohort study during an epidemic in Quebec.
Clin Infect Dis. 2005;41(9):1254-1260.
CHAPTER 49

Gastric Helicobacter-like Infections


Jane E. Sykes and Stanley L. Marks

for development of gastric adenocarcinoma and lymphoma. In


Overview of Helicobacter Infections in Dogs contrast, the extent to which Helicobacter spp. cause disease
and Cats in dogs and cats is not fully understood, and newly discovered
First Described: 1881 by Rappin (France),1 who observed organisms in dogs and cats have drawn most attention in regard
­spiral bacteria in a dog’s stomach; the association to their zoonotic potential.5
between H. pylori infection and disease in humans was Helicobacter pylori, the type organism, is the most important
made in 1983 by Warren and Marshall (Australia).2 species infecting humans. H. pylori infection is extremely rare in
cats and has only been identified in one colony of cats to date.6
Cause: Various species of non–H. pylori helicobacters, rarely
It has not been described in dogs. Dogs and cats are generally
H. pylori in cats (gram-negative spiral-shaped bacteria
infected with gastric non–H. pylori helicobacters (also referred
that belong to the family Helicobacteraceae)
to as gastric helicobacter-like organisms, or GHLOs).5 These
Primary Mode of Transmission: Fecal-oral and oral-oral trans- are much larger than H. pylori (5 to 10 µm long versus 1.5 to
mission proposed, possibly water-borne. Transmission 3 µm long for H. pylori), and species cannot be differentiated
through contact with vomitus may also occur. from one another based on their light or electron microscopic
Affected Hosts: Humans and a variety of other animals are appearance alone (Figure 49-1). The nomenclature of non–H.
colonized by helicobacters; an association with disease is pylori helicobacters is complicated. Species identified in dogs
clearest for humans and ferrets and cats are listed in Table 49-1.4,7-16 Before their genetic charac-
terization, non–H. pylori helicobacters were originally referred
Geographic Distribution: Worldwide
to as ­“Gastrospirillum hominis” and later “Helicobacter heil-
Major Clinical Signs (Dogs and Cats): Possibly chronic vomiting mannii.” Subsequent analysis of multiple gene sequences from
(gastric helicobacters) or diarrhea (intestinal helicobacters), a variety of “H. heilmannii” from dogs and cats has revealed
although evidence for a causative role in disease is weak that “H. heilmannii” is not one but a group of organisms that
Differential Diagnoses: Dietary indiscretion, gastrointestinal includes H. felis, H. bizzozeronii, H. salomonis, and an organ-
foreign body, chronic pancreatitis, inflammatory bowel dis- ism that has been confusingly named H. heilmannii. These
ease, food-responsive enteropathy, eosinophilic fibrosing organisms have also been implicated in human disease, albeit
gastritis (cats), bilious vomiting syndrome, gastrointestinal less commonly than H. pylori.5
neoplasia (lymphoma, mast cell neoplasia, gastric adeno- Depending on the study, between 60% and 100% of dogs
carcinoma), chronic infiltrative infectious diseases of the and cats carry non–H. pylori helicobacters, and gastric Heli-
gastrointestinal tract (including feline infectious peritonitis, cobacter spp. can be found in apparently healthy animals and
mycobacteriosis, canine cryptococcosis, pythiosis), hypo- dogs and cats without signs of vomiting.5,17-22 H. felis infection
adrenocorticism, hyperthyroidism, toxins (including drugs), has been associated with gastric pathology in dogs in some
gastric helminthiasis (e.g., P
­ hysaloptera and Ollulanus spp.) studies23 but not others.24 Dogs and cats are probably colo-
nized shortly after birth, as a result of fecal-oral and oral-oral
Human Health Significance: Dogs and cats may be a source ­transmission.25 Transmission through contact with vomitus may
of human infection with non–H. pylori helicobacters that also occur. Water-borne transmission may play a role in spread of
have been associated with gastritis, gastroduodenal H. pylori to humans.26 Shelter and colony dogs and cats may
ulceration, and low-grade mucosa-associated lymphoid have a higher prevalence of colonization, most likely due to the
tissue (MALT) lymphoma in humans. close ­proximity of animals to one another. One Helicobacter
species may be able to suppress the presence of another, so that
dogs and cats are primarily colonized with a single species,
although in some animals, the simultaneous presence of mul-
Etiology and Epidemiology tiple species has been found.27-30 The organisms have a remark-
able ability to survive the low pH of the stomach, which they
Helicobacter spp. are flagellate, gram-negative, microaerophilic, resist by living deep in the mucus glands of the stomach and
curved to spiral-shaped motile bacteria. They are grouped through production of the enzyme urease. The urease catalyzes
into gastric, hepatic, and intestinal Helicobacter species, with the hydrolysis of urea to carbon dioxide and ammonia, which
intestinal species residing primarily in the large intestine.3,4 In raises the pH of the organism’s milieu. In dogs and cats, the
humans, gastric Helicobacter spp. are an important cause of organisms are also found within the canaliculi and the cyto-
gastritis and gastroduodenal ulceration and increase the risk plasm of parietal cells.25,30,31

465
466 SECTION 2  Bacterial Diseases

concurrently.31 Experimental and natural infection of dogs and


cats with non–H. pylori helicobacters has been associated with
chronic lymphoplasmacytic gastritis and lymphoid follicular
hyperplasia. Gastroduodenal ulceration and marked altera-
tions in the gastric acid secretory axis, as occur in humans,
have not been observed in dogs and cats.24,33,34 However, not
all animals infected with non–H. pylori helicobacters have his-
topathologic evidence of gastritis, and no correlation has been
observed between the severity of gastric pathology and the
degree of colonization by helicobacters. More severe gastritis
with marked lymphoid follicular hyperplasia and neutrophilic
inflammatory infiltrates has been reported in cats and acutely in
puppies experimentally infected with H. pylori.35,36 The puppies
developed gastrointestinal signs, including vomiting and loose
stool, shortly after inoculation. It is possible that the degree of
bacterial invasion and gastritis in dogs and cats varies with the
­ elicobacter species or even strain.30 In humans, gas-
infecting H
tric pathology has been associated with possession of a number
of pathogenicity genes by H. pylori, including cagA (cytotoxin-
associated gene A), vacA (vacuolating cytotoxin), and iceA
A B (induced by contact with epithelium).37 The degree of vacuolat-
FIGURE 49-1  Structure of gastric Helicobacter species. Note the terminal bunches of ing cytotoxin production can also vary among strains.38
flagellar filaments. A, Helicobacter pylori. B, Gastric helicobacter-like organism. In human patients, H. pylori infection has been well associ-
ated with gastric low-grade, B-cell, mucosa-associated lymphoid
tissue (MALT) lymphoma. Eradication of H. pylori achieves
complete remission in some patients with H. pylori–positive
TABLE 49-1 early-stage gastric MALT lymphoma.39 One study suggested
a possible association between Helicobacter spp. infection and
Helicobacter Species Identified in Dogs, Cats, and Humans4,7-16 gastric lymphoma in cats,40 but additional studies are required
to determine the role of Helicobacter spp. infection in feline gas-
Species Location Host tric disease.
H. bizzozeronii Stomach Dog, humans Non–H. pylori helicobacters have been detected in the hep-
H. salomonis Stomach Dog atobiliary system of cats with neutrophilic cholangitis41 and
H. cynogastricus Stomach Dog lymphocytic cholangitis,42 although this may reflect ascending
H. heilmannii Stomach Dog, cat, humans bacterial infection secondary to an underlying disease process.
H. felis Stomach Dog, cat, humans H. canis was detected in the liver of a 2-month-old puppy with
H. pametensis Stomach Cat multifocal necrotizing hepatitis that had acute weakness and
H. baculiformis Stomach Cat vomiting, and died within hours.43 H. canis was also detected in
H. pylori Stomach Cat a colony of Bengal cats with diarrhea, but its role as a causative
“Flexispira rappini” Stomach, intestine Dog, cat agent was uncertain.44 Abundant organisms that genetically
H. bilis Stomach, intestine Dog, cat resembled H. canis were detected microscopically throughout
H. cinaedi Intestine Dog, cat the large intestine of a 2-month-old kitten with severe diarrhea,
H. fennelliae Intestine Dog, cat vomiting, dehydration, weight loss, and inappetence.3 In one
“H. colifelis” Intestine Cat study, the presence of heavy colonic infection with enterohe-
H. marmotae Intestine Cat patic Helicobacter spp. infection in a group of laboratory dogs
H. canis Intestine, liver Dog, cat, humans was associated with the presence of mucosal atrophy and fibro-
sis, and a possible role of Helicobacter spp. infection in canine
inflammatory bowel disease was suggested.29

Clinical Features Diagnosis


Signs and Their Pathogenesis Microbiologic Testing
The majority of dogs and cats infected with Helicobacter spp. Clinical diagnosis of Helicobacter infection in dogs and cats has
show no clinical signs. Helicobacter infection of some dogs most commonly been based on histopathology, cytology, rapid
and cats has been hypothesized to cause chronic intermittent urease testing, and, to a lesser extent, PCR on gastric biopsies
vomiting, inappetence, pica, belching, weight loss, fever, and (Table 49-2). Other tests such as the urea breath and blood
polyphagia.31 Unfortunately, strong evidence that supports tests, fecal PCR, and serologic tests are primarily used in human
an association with such disease in dogs and cats is lacking. patients and in the research arena.
Clinical signs in some animals with biopsy-confirmed infec-
tion resolve following specific therapy for gastric helicobacter Diagnosis Using Cytology and Histopathology
infection,31-33 although the results of one study were difficult Spiral organisms can be readily detected in touch impression
to interpret because an elimination diet was administered smears of gastric tissue obtained following biopsy or necropsy,
CHAPTER 49  Gastric Helicobacter-like Infections 467

TABLE 49-2
Diagnostic Assays Available for Gastric Helicobacter Infection in Dogs and Cats*
Assay Specimen Type Target Performance
Bacterial isolation Gastric biopsies Helicobacter Can take up to 10 days. Low sensitivity for non–H. pylori
organisms helicobacters, which can be difficult to culture. Patchy
distribution may also lead to false negatives. Allows
­typing and antimicrobial susceptibility testing.
Cytology Impression smears Helicobacter Rapid and sensitive, but false negatives can occur.
of ­gastric biopsies organisms Patchy distribution may also lead to false negatives.
or ­cytology brush Does not provide information on inflammatory or
­specimens architectural changes in underlying tissue.
Histopathology Gastric biopsies Helicobacter Less sensitive than cytologic examination. Patchy
organisms ­distribution may also lead to false negatives. Use
of silver stains increases sensitivity when organism
­numbers are low. ­Allows determination of associated
gastric ­histopathology.
Rapid urease Gastric biopsies Helicobacter Rapid and very sensitive. Patchy distribution and low
testing urease ­organism loads may lead to false negatives. Very rare
false positives with other urease-producing organisms.
PCR Gastric biopsies Helicobacter DNA Rapid (within hours); can be highly sensitive and
specific. Sequence analysis can determine the species
involved. Primarily used on a research basis.
Serology Serum Antibodies to Moderate sensitivity in dogs and cats. Results do not
Helicobacter spp. ­correlate well with gastric histopathology and do not
­always correlate with active infection. Primarily used
in research settings.
Urea breath and Breath, blood Radiolabeled carbon Requires specialized detection equipment. Although
blood testing dioxide derived from ­sensitive, false negatives may occur, such as following
the activity of acid suppression. In dogs and cats, has been primarily
Helicobacter urease used in research settings to date.

*For all tests, positive results do not imply that Helicobacter is the cause of disease.

after staining with Gram or Diff Quik stains (Figure 49-2). A been documented in non–H. pylori helicobacters isolated from
cytology brush can also be used to obtain a specimen during dogs and cats.48
endoscopy. Several biopsies or brush specimens may need to be
evaluated from different regions of the stomach (fundus, cardia, Rapid Urease Testing
antrum/pylorus) when the distribution of organisms is patchy. The rapid urease test involves incubating a gastric biopsy in a
When organism burdens are high, the organisms can be detected urea broth that contains the pH indicator phenol red. If gastric
using histopathology, which allows assessment of concurrent helicobacters are present, helicobacter urease breaks down the
gastric pathology (Figures 49-3 and 49-4). The use of silver urea; with the release of ammonia, a rise in pH and a color
stains (such as Warthin-Starry stain or Steiner stain), Giemsa, change occur. The change in color can occur within 1 to 3 hours,
or toluidine blue stain, as well as immunostaining, dramatically although the broth is generally incubated at room temperature
increases sensitivity for organism detection (see Figures 49-3, C, for 24 hours. The test is very sensitive, but false-negative results
and 49-4, C).45 can occur if the distribution within the stomach is patchy or if
organism loads are low. False-positive results have rarely been
Bacterial Isolation reported when other urease-producing bacteria are present in
Isolation of gastric helicobacters is difficult and has low sen- the stomach, such as Proteus spp.
sitivity. Growth typically requires incubation on selective
media in microaerobic conditions for 5 to 10 days. Isolation is Detection of Helicobacter DNA
advantageous in research settings because it allows subsequent PCR assays have been extensively used to detect Helicobacter
identification of the organism using conventional biochemical species in tissues on a research basis, including those from dogs
testing, whole-cell protein profiling, and DNA analysis.21,46 and cats. Sequence analysis of the PCR products can allow iden-
It also allows antimicrobial susceptibility testing, which tification of the Helicobacter species present. Fluorescent in
has recently been recommended before treating humans for situ hybridization (FISH) has also been used to detect, localize,
H. pylori infection because of the increasing prevalence of anti- and identify Helicobacter species infection in tissues from dogs
microbial resistance.47 Resistance to antimicrobials has also and cats.4,32,40,49
468 SECTION 2  Bacterial Diseases

FIGURE 49-2  Spiral-shaped gastric Helicobacter-like organisms visible in a Diff Quik–stained smear (arrows) of a gastric biopsy from a 2-year-old male neutered Cavalier King Charles
spaniel with chronic vomiting and regurgitation for which no other cause was apparent (1000× magnification).

Serology some cats.58 Neutrophil and eosinophil infiltrates can accom-


A variety of serum ELISA and immunoblotting assays have pany mononuclear inflammation in some cats infected with
been used in humans to detect antibodies and screen for H. pylori.58,60 Organisms may be found in the ­superficial
H. pylori infection, and some can be used to some extent to mucus and crypts, and sometimes within parietal cells see
monitor the success of therapy.50,51 Serologic assays have been Figures 49-3 and 49-4).
used on a research basis to noninvasively detect non–H. pylori Histopathologic findings in a cat infected with an intestinal
­helicobacter infection in dogs and cats, in some studies demon- helicobacter included large numbers of densely packed spiral
strating ­moderate sensitivity and good specificity.52-54 bacteria covering the intestinal mucosa and present within the
crypts, and minimal inflammatory changes. Hepatic changes
Urea Breath and Blood Testing in a dog with H. canis infection consisted of randomly distrib-
Diagnosis and monitoring of the extent of H. pylori infec- uted and coalescing hepatocellular necrosis with associated
tion in human patients can also be accomplished noninva- ­neutrophilic and mononuclear infiltrates.43
sively using urea breath and blood testing, which involves oral
administration of urea containing 14C or the stable isotope 13C. Treatment and Prognosis
After ingestion, the labeled urea is converted to ammonia by
­Helicobacter urease, and the released carbon is absorbed sys- Whether antimicrobial treatment should be used for
temically, where it can be measured in the blood. Subsequently, ­Helicobacter infections in dogs and cats is unknown.
the labeled carbon dioxide is exhaled and can be measured Humans with H. pylori infections are typically treated with
in the breath. Acid suppression interferes with test results by a ­combination of a proton pump inhibitor, amoxicillin, and
decreasing H
­ elicobacter urease activity, and so testing is gener- clarithromycin, with or without bismuth subsalicylate. Treat-
ally performed several days after discontinuing treatment. False ment of humans is reserved for symptomatic individuals; the
positives can also occur.55 Urea breath and blood testing has development of antimicrobial resistance by H. pylori is increas-
been used with success in dogs,56 and breath testing has been ingly of concern.61 Treatment should be reserved for dogs and
used in cats20,57 both before and following specific treatment for cats with gastrointestinal signs that have no other identifiable
Helicobacter infection. cause of illness, after a diagnosis of gastritis and Helicobacter
infection has been confirmed using biopsy. A 2-week course
Stool Antigen Assays of combination therapy with ­antimicrobials and a proton
Assays that detect H. pylori antigen in feces have been used in pump inhibitor or an H2 antagonist does not reliably eliminate
human patients,55 but their use in dogs and cats has not been gastric helicobacters in dogs and cats, although clinical signs
described. often resolve.33,56,62,63 Reinfection can occur in some animals
after treatment. This theory is underscored by a study of 20
Pathologic Findings dogs that were naturally infected with Helicobacter spp. and
Infection with non–H. pylori helicobacters in dogs and cats has treated with triple therapy ­(clarithromycin, amoxicillin, and
primarily been associated with chronic lymphocytic or lym- lansoprazole) for 7 days. The dogs were then randomized into
phoplasmacytic gastritis, with lymphoid follicular hyperplasia a control group kept in isolation, and an experimental group,
(see Figures 49-3, A, and 49-4, A), although the extent of which was placed in contact with Helicobacter-positive dogs
inflammation varies dramatically from absent to severe.19,58,59 for 60 days. Triple therapy was effective in 100% of the dogs;
An eosinophilic inflammatory component has been described in however, recurrence of infection occurred in 80% of dogs in
CHAPTER 49  Gastric Helicobacter-like Infections 469

A A

B B

C C
FIGURE 49-3  Histopathology of the stomach of a 2-year-old female spayed Mal- FIGURE 49-4  Histopathology of the stomach of a 14-year-old male neutered
tese terrier with chronic vomiting and regurgitation. A, Lymphoid follicular nodules. The domestic shorthair that was infected with FIV and was euthanized because of squamous
remaining lamina propria is infiltrated by small numbers of eosinophils, neutrophils, lym- cell carcinoma. Gastrointestinal signs were not reported. A, Severe, chronic, lymphofol-
phocytes, and plasma cells. H&E stain, 40× magnification. B, Large numbers of spiral- licular gastritis. H&E stain, 40× magnification. B, Abundant fine, spiral-shaped bacteria
shaped bacteria are visible in the superficial mucous and gastric glands. H&E stain, 1000× were visualized in the superficial mucous and within parietal cells (arrows). H&E stain.
magnification. C, Agyrophilic intralesional spiral-shaped bacteria within the gastric C, Warthin-Starry stain showing intracellular agyrophilic spiral-shaped bacteria (arrows);
glands. Warthin-Starry stain, 1000× magnification. 1000x magnification.
470 SECTION 2  Bacterial Diseases

TABLE 49-3
Antimicrobials Used with Success to Treat Helicobacter spp.
Infection in Dogs and Cats
Dose Interval Duration
Drug (mg/kg) Route (hours) (days)
Metronidazole 10-15 PO 12 14-21
Amoxicillin 22 PO 12 14-21
Bismuth 0.22 mL/kg PO 6-8 14-21
subsalicylate
Clarithromycin 5-10 PO 12 14-21
Omeprazole 0.7-1.0 PO 24 14-21
Famotidine 0.5-1.0 PO 12 14-21

The first three drugs are administered as a combination. Alternatively, clar-


ithromycin can be combined with amoxicillin and administered with either
omeprazole or famotidine. Bismuth compounds should be avoided in cats.
Omeprazole should be given on an empty stomach 30 minutes before a meal. FIGURE 49-5  Endoscopic image of lymphoid follicular gastritis in a 2-year-old
­maltese terrier with chronic vomiting and regurgitation that was associated with the
the experimental group, and in none of the dogs in the con- presence of large numbers of gastric helicobacter like-organisms. Note the ‘goose pimple’
trol group after 60 days.64 A 3-week course of amoxicillin, appearance of the mucosa.
metronidazole, and bismuth subsalicylate (Table 49-3) cleared
gastric helicobacters in dogs and cats, with resolution of vomit- the organism, as well as the possession of virulence factors by
ing, but histopathologic evidence of gastritis did not resolve.32 H. pylori strains, appears to play an important role.66 Eradication
Metronidazole could be substituted with clarithromycin.21 One of infection using antimicrobial therapy can cure gastroduode-
study showed that concurrent use of an H2 antagonist in dogs nal ulceration and MALT lymphoma, but gastric adenocarci-
did not change outcome.33 This is consistent with the lack of noma generally persists despite treatment.67 Although H. pylori
abnormalities in the gastric acid secretory axis in dogs infected has been documented to infect a colony of cats, infection of cats
with non–H. pylori helicobacters.24,34 with this species is extremely rare, and exposure to pets is not a
risk factor for H. pylori infection in humans.
Immunity and Vaccination Non–H. pylori gastric helicobacter infection is uncom-
mon in humans compared with H. pylori infection. Never-
Gastric helicobacters persist in the stomach despite a vigorous theless, these helicobacters have also been associated with
humoral immune response. Research has been underway chronic active gastritis, gastroduodenal ulceration, and
to develop an effective vaccine for H. pylori infection in MALT ­lymphomas in people. Disease is generally less severe
humans, and a variety of H. pylori antigens induce protection than that caused by H. pylori.5 Clinical signs in humans may
in healthy human volunteers or mouse models.65 No vaccine be absent or consist of nausea, epigastric pain, vomiting,
is available for Helicobacter infection in dogs and cats, and hematemesis, heartburn, and decreased appetite. Treatment
a greater understanding of the role of gastric helicobacters in with triple antibiotic therapy and bismuth subsalicylate can
canine and feline disease will be required in order to assess resolve infection and gastritis. Pets have been suggested as
the need for effective vaccines against these organisms. a source of these helicobacters for humans,5,7,68-71 and close
contact with dogs and cats is a risk factor for infection.72
Public Health Aspects Licking has been suggested as a mode of transmission, based
on the presence of non–H. pylori helicobacter DNA in the
At least 50% of the world’s human population is infected with oral cavity of dogs.25 Pigs play an even more important role
H. pylori, although the prevalence of infection has decreased in zoonotic ­transmission of non–H. pylori gastric helicobac-
in developed countries.66 In the Western world, 1% to 10% of ters, the swine helicobacter H. suis being the most prevalent
people develop gastroduodenal ulceration as a result of chronic ­species ­identified in humans.5
H. pylori infection. A smaller percentage of infected people Intestinal helicobacters found in dogs and cats, especially
(<3%) develop gastric adenocarcinoma or MALT lymphoma H. cinaedi, have been associated with enteric disease and
in association with infection. Duodenal ulceration results from ­bacteremia in immunocompromised humans.73 The relative role
increased gastrin release in response to H. pylori–induced of dogs and cats in transmission of these organisms to humans
­gastric antral inflammation. Factors that influence outcome are is unknown. Nosocomial spread of H. cinaedi was suggested
not well understood, but an individual’s immune response to among immunocompromised humans in one report.74
CHAPTER 49  Gastric Helicobacter-like Infections 471

CASE EXAMPLE BUN 22 mg/dL (5-21 mg/dL)


Creatinine 1.1 mg/dL (0.3-1.2 mg/dL)
Glucose 95 mg/dL (64-123 mg/dL)
Signalment: “Mochi,” a 2-year-old female spayed Maltese Total protein 5.7 g/dL (5.4-7.6 g/dL)
terrier from Dixon, CA Albumin 4.0 g/dL (3.0-4.4 g/dL)
History: Mochi had been vomiting and regurgitating Globulin 1.7 g/dL (1.8-3.9 g/dL)
intermittently since she was 1 year of age. As a puppy, ALT 42 U/L (19-67 U/L)
the owner (a veterinary student) reported intermittent AST 45 U/L (19-42 U/L)
episodes of “burping.” The vomiting occurred 2 to 5 times ALP 52 U/L (21-170 U/L)
throughout the day and was not associated with eating, GGT < 3 U/L (0-6 U/L)
drinking, or exercise. The vomitus usually consisted of brown Cholesterol 241 mg/dL (135-361 mg/dL)
fluid with no recognizable food particles. Dietary changes Total bilirubin 0.2 mg/dL (0-0.2 mg/dL).
that included a digestible elimination diet containing a Urinalysis: SGr 1.040, pH 7.0, no protein, glucose, or ketones,
novel, single protein source followed by a fat-restricted trace hemoprotein, 0-1 WBC/HPF, 0-1 RBC/HPF, moderate
prescription diet had not resulted in improvement in her lipid droplets.
clinical signs. Therapy with cisapride (3 mg PO q8h) and Serum Pancreatic Lipase Immunoreactivity: <30 µg/L
famotidine (2.5 mg PO q24h) or omeprazole (3.5 mg PO (reference range 0-200 µg/L)
q24h) resulted in only slight improvement in the frequency Imaging Findings:
of vomiting. Her appetite was appropriate, and there had Abdominal Radiographs: A moderate amount of gas was
been no weight loss or diarrhea. She primarily lived indoors identified within multiple small intestinal loops, but an
with one cat. She was up to date on vaccinations, which obstructive pattern was not identified.
included those for rabies, distemper, parvovirus, and Thoracic Radiographs: The cardiopulmonary structures were
canine adenovirus. within normal limits. A small amount of gas was visualized
Physical Examination: within the esophagus on the left lateral projections.
Body Weight: 3 kg Esophagram: Unremarkable study.
General: Bright, alert and responsive, hydrated, nervous. Abdominal Ultrasound: No abnormalities were identified.
Ambulatory on all 4 limbs. T = 100.4°F (38.0°C), HR = 120 Gastroduodenoscopy: Gross findings were suggestive
beats/min, RR = 28 breaths/min, mucous membranes pink of diffuse lymphoid follicular hyperplasia, with a mild
and moist, CRT = 1 s. “goose pimple” appearance to the gastric mucosa
Integument, Eyes, Ears, Nose, and Throat: No clinically (Figure 49-5). Multiple biopsies of all regions of the stomach
significant abnormalities were noted. and multiple duodenal biopsies were obtained and
Musculoskeletal: The dog’s body condition score was 4/9 with submitted for histopathology.
symmetrical muscling. Histopathology: Stomach: Moderate, lymphofollicular,
Cardiovascular, Respiratory, and Lymph Nodes: No eosinophilic, and lymphoplasmacytic gastritis with intralesional
clinically significant abnormalities were noted. spiral bacteria (see Figure 49-3). Duodenum: Mild, chronic,
Gastrointestinal and Genitourinary: Abdominal palpation lymphofollicular, eosinophilic, and lymphoplasmacytic
was unremarkable. “Burping” behavior was observed in duodenitis and ileitis. Jejunum: Diffuse, mild, eosinophilic, and
the room, which occurred without any prodromal signs lymphoplasmacytic enteritis.
or abdominal effort. Lip-smacking behavior occurred Diagnosis: Chronic gastritis associated with Helicobacter
immediately afterwards. Rectal examination was normal. spp. infection; mild eosinophilic and lymphoplasmacytic
Laboratory Findings: enteritis.
CBC: Treatment: Treatment for the Helicobacter infection was
HCT 46.5% (40%-55%) initiated with metronidazole (30 mg PO q12h), amoxicillin
MCV 64.4 fL (65-75 fL) (60 mg PO q12h), and bismuth subsalicylate for 2 weeks.
MCHC 35.9 g/dL (33-36 g/dL) The owner reported almost complete resolution of the
WBC 5700 cells/µL (6000-13,000 cells/µL) vomiting following treatment, but signs returned after
Neutrophils 3112 cells/µL (3000-10,500 cells/µL) treatment was discontinued, and an additional course of
Lymphocytes 1830 cells/µL (1000-4000 cells/µL) treatment was instituted. Treatment with a fat-restricted
Monocytes 257 cells/µL (150-1200 cells/µL) prescription diet was continued. The frequency of vomiting
Eosinophils 479 cells/µL (0-1500 cells/µL) then decreased to once a week.
Platelets 277,000/µL (150,000-400,000 platelets/µL). Comments: Whether the Helicobacter spp. infection in this dog
Serum Chemistry Profile: contributed to clinical signs or was an incidental finding in
Sodium 147 mmol/L (145-154 mmol/L) a dog with underlying enteritis was not clear. The failure to
Potassium 4.4 mmol/L (3.6-5.3 mmol/L) respond to multiple diet changes, including an elimination
Chloride 111 mmol/L (108-118 mmol/L) diet, and the dramatic response to triple therapy supported
Bicarbonate 23 mmol/L (16-26 mmol/L) a possible role for Helicobacter spp. infection, or a diagnosis of
Phosphorus 5.3 mg/dL (3.0-6.2 mg/dL) antibiotic-responsive gastroenteropathy that was unrelated
Calcium 10.6 mg/dL (9.7-11.5 mg/dl) to Helicobacter spp. infection.
472 SECTION 2  Bacterial Diseases

SUGGESTED READING 20. Neiger R, Dieterich C, Burnens A, et  al. Detection and
­prevalence of Helicobacter infection in pet cats. J Clin Microbiol.
Haesebrouck F, Pasmans F, Flahou B, et  al. Gastric helicobacters in 1998;36:634-637.
domestic animals and nonhuman primates and their significance for 21. Neiger R, Simpson KW. Helicobacter infection in dogs and cats:
human health. Clin Microbiol Rev. 2009;22(2):202-223. facts and fiction. J Vet Intern Med. 2000;14:125-133.
22. Otto G, Hazell SH, Fox JG, et al. Animal and public health implica-
REFERENCES tions of gastric colonization of cats by helicobacter-like organisms.
1. Rappin JP. Contre l’etude de bacterium de la bouche etat normal. J Clin Microbiol. 1994;32:1043-1049.
In: College de France, Nantes; 1881. 23. Lee A, Krakowka S, Fox JG, et  al. Role of Helicobacter felis in
2. Warren JR, Marshall BJ. Unidentified curved bacilli on gastric chronic canine gastritis. Vet Pathol. 1992;29:487-494.
­epithelium in active chronic gastritis. Lancet. 1983;1:1273-1275. 24. Simpson KW, McDonough PL, Strauss-Ayali D, et al. Helicobacter
3. Foley JE, Solnick JV, Lapointe JM, et al. Identification of a novel felis infection in dogs: effect on gastric structure and function. Vet
enteric Helicobacter species in a kitten with severe diarrhea. J Clin Pathol. 1999;36:237-248.
Microbiol. 1998;36:908-912. 25. Recordati C, Gualdi V, Tosi S, et al. Detection of Helicobacter spp.
4. Recordati C, Gualdi V, Craven M, et  al. Spatial distribution of DNA in the oral cavity of dogs. Vet Microbiol. 2007;119:346-351.
Helicobacter spp. in the gastrointestinal tract of dogs. ­Helicobacter. 26. Travis PB, Goodman KJ, O’Rourke KM, et  al. The association
2009;14:180-191. of drinking water quality and sewage disposal with Helicobacter
5. Haesebrouck F, Pasmans F, Flahou B, et al. Gastric helicobacters pylori incidence in infants: the potential role of water-borne
in domestic animals and nonhuman primates and their significance ­transmission. J Water Health. 2010;8:192-203.
for human health. Clin Microbiol Rev. 2009;22:202-223. 27. Wiinberg B, Spohr A, Dietz HH, et  al. Quantitative analysis of
6. Handt LK, Fox JG, Dewhirst FE, et al. Helicobacter pylori isolated inflammatory and immune responses in dogs with gastritis and
from the domestic cat: public health implications. Infect Immun. their relationship to Helicobacter spp. infection. J Vet Intern Med.
1994;62:2367-2374. 2005;19:4-14.
7. Alon D, Paitan Y, Ben-Nissan Y, et  al. Persistent Helicobacter 28. Ekman E, Fredriksson M, Trowald-Wigh G, et  al. Helicobacter
canis bacteremia in a patient with gastric lymphoma. Infection. spp. in the saliva, stomach, duodenum and faeces of colony dogs.
2010;38:62-64. Vet J. 2013;195:127-129.
8. Baele M, Decostere A, Vandamme P, et al. Helicobacter baculifor- 29. Castiglioni V, Vailati Facchini R, Mattiello S, et al. ­Enterohepatic
mis sp. nov., isolated from feline stomach mucosa. Int J Syst Evol Helicobacter spp. in colonic biopsies of dogs: molecular,
Microbiol. 2008;58:357-364. ­histopathological and immunohistochemical investigations. Vet
9. De Groote D, Van Doorn LJ, Van den Bulck K, et  al. Detection Microbiol. 2012;159:107-114.
of non-pylori Helicobacter species in “Helicobacter heilmannii”- 30. Lanzoni A, Faustinelli I, Cristofori P, et  al. Localization of Heli-
infected humans. Helicobacter. 2005;10:398-406. cobacter spp. in the fundic mucosa of laboratory Beagle dogs: an
10. Fox JG, Shen Z, Xu S, et  al. Helicobacter marmotae sp. nov. ultrastructural study. Vet Res. 2011;42:42.
isolated from livers of woodchucks and intestines of cats. J Clin 31. Lecoindre P, Chevallier M, Peyrol S, et al. Gastric helicobacters in
Microbiol. 2002;40:2513-2519. cats. J Feline Med Surg. 2000;2:19-27.
11. Hanninen ML, Happonen I, Saari S, et al. Culture and ­characteristics 32. Jergens AE, Pressel M, Crandell J, et  al. Fluorescence in situ
of Helicobacter bizzozeronii, a new canine gastric Helicobacter hybridization confirms clearance of visible Helicobacter spp.
sp. Int J Syst Bacteriol. 1996;46:160-166. associated with gastritis in dogs and cats. J Vet Intern Med.
12. Jalava K, Kaartinen M, Utriainen M, et al. Helicobacter ­salomonis 2009;23:16-23.
sp. nov., a canine gastric Helicobacter sp. related to H ­ elicobacter 33. Leib MS, Duncan RB, Ward DL. Triple antimicrobial therapy
felis and Helicobacter bizzozeronii. Int J Syst Bacteriol. 1997; and acid suppression in dogs with chronic vomiting and gastric
47:975-982. ­Helicobacter spp. J Vet Intern Med. 2007;21:1185-1192.
13. Kiehlbauch JA, Brenner DJ, Cameron DN, et  al. Genotypic and 34. Simpson KW, Strauss-Ayali D, McDonough PL, et  al. Gastric
phenotypic characterization of Helicobacter cinaedi and Helico- function in dogs with naturally acquired gastric Helicobacter spp.
bacter fennelliae strains isolated from humans and animals. J Clin ­infection. J Vet Intern Med. 1999;13:507-515.
Microbiol. 1995;33:2940-2947. 35. Perkins SE, Fox JG, Marini RP, et al. Experimental infection in cats
14. Kivisto R, Linros J, Rossi M, et  al. Characterization of multiple with a cagA+ human isolate of Helicobacter pylori. Helicobacter.
Helicobacter bizzozeronii isolates from a Finnish patient with 1998;3:225-235.
severe dyspeptic symptoms and chronic active gastritis. Helico- 36. Rossi G, Rossi M, Vitali CG, et  al. A conventional beagle dog
bacter. 2010;15:58-66. model for acute and chronic infection with Helicobacter pylori.
15. Rossi M, Hanninen ML, Revez J, et al. Occurrence and species level Infect Immun. 1999;67:3112-3120.
diagnostics of Campylobacter spp., enteric Helicobacter spp. and 37. Ramis IB, Fonseca TL, de Moraes EP, et  al. Molecular basis of
Anaerobiospirillum spp. in healthy and diarrheic dogs and cats. Vet pathogenicity in Helicobacter pylori clinical isolates. J Clin
Microbiol. 2008;129:304-314. ­Microbiol. 2010;48:3776-3778.
16. Van den Bulck K, Decostere A, Baele M, et al. Helicobacter cyno- 38. Atherton JC, Cao P, Peek Jr RM, et al. Mosaicism in ­vacuolating
gastricus sp. nov., isolated from the canine gastric mucosa. Int J cytotoxin alleles of Helicobacter pylori. Association of specific
Syst Evol Microbiol. 2006;56:1559-1564. vacA types with cytotoxin production and peptic ulceration. J Biol
17. Eaton KA, Dewhirst FE, Paster BJ, et  al. Prevalence and variet- Chem. 1995;270:17771-17777.
ies of Helicobacter species in dogs from random sources and pet 39. Chen LT, Lin JT, Tai JJ, et al. Long-term results of ­anti–Helicobacter
dogs: animal and public health implications. J Clin Microbiol. pylori therapy in early-stage gastric high-grade transformed
1996;34:3165-3170. MALT lymphoma. J Natl Cancer Inst. 2005;97:1345-1353.
18. Happonen I, Linden J, Saari S, et al. Detection and effects of heli- 40. Bridgeford EC, Marini RP, Feng Y, et  al. Gastric Helicobacter
cobacters in healthy dogs and dogs with signs of gastritis. J Am Vet ­species as a cause of feline gastric lymphoma: a viable hypothesis.
Med Assoc. 1998;213:1767-1774. Vet Immunol Immunopathol. 2008;123:106-113.
19. Hermanns W, Kregel K, Breuer W, et al. Helicobacter-like organ- 41. Greiter-Wilke A, Scanziani E, Soldati S, et  al. Association of
isms: histopathological examination of gastric biopsies from dogs Helicobacter with cholangiohepatitis in cats. J Vet Intern Med.
and cats. J Comp Pathol. 1995;112:307-318. 2006;20:822-827.
CHAPTER 49  Gastric Helicobacter-like Infections 473

42. Otte CMA, Perez Gutierrez O, Favier RP, et  al. Detection of 58. Scanziani E, Simpson KW, Monestiroli S, et  al. Histological and
bacterial DNA in bile of cats with lymphocytic cholangitis. Vet immunohistochemical detection of different Helicobacter species in
­Microbiol. 2012;156:217-221. the gastric mucosa of cats. J Vet Diagn Invest. 2001;13:3-12.
43. Fox JG, Drolet R, Higgins R, et al. Helicobacter canis isolated from 59. Simpson KW, Strauss-Ayali D, Scanziani E, et al. Helicobacter felis
a dog liver with multifocal necrotizing hepatitis. J Clin Microbiol. infection is associated with lymphoid follicular hyperplasia and
1996;34:2479-2482. mild gastritis but normal gastric secretory function in cats. Infect
44. Foley JE, Marks SL, Munson L, et  al. Isolation of Helicobacter Immun. 2000;68:779-790.
canis from a colony of Bengal cats with endemic diarrhea. J Clin 60. Simpson KW, Strauss-Ayali D, Straubinger RK, et al. Helicobacter
Microbiol. 1999;37:3271-3275. pylori infection in the cat: evaluation of gastric colonization,
45. Prachasilpchai W, Nuanualsuwan S, Chatsuwan T, et al. ­Diagnosis inflammation and function. Helicobacter. 2001;6:1-14.
of Helicobacter spp. infection in canine stomach. J Vet Sci. 61. Graham DY, Fischbach L. Helicobacter pylori treatment in the era
2007;8:139-145. of increasing antibiotic resistance. Gut. 2010;59:1143-1153.
46. Jalava K, On SL, Vandamme PA, et al. Isolation and identification 62. Happonen I, Linden J, Westermarck E. Effect of triple therapy
of Helicobacter spp. from canine and feline gastric mucosa. Appl on eradication of canine gastric helicobacters and gastric disease.
Environ Microbiol. 1998;64:3998-4006. J Small Anim Pract. 2000;41:1-6.
47. Wenzhen Y, Yumin L, Quanlin G, et al. Is antimicrobial suscepti- 63. Perkins SE, Yan LL, Shen Z, et  al. Use of PCR and culture to
bility testing necessary before first-line treatment for Helicobacter detect Helicobacter pylori in naturally infected cats following
pylori infection? Meta-analysis of randomized controlled trials. triple antimicrobial therapy. Antimicrob Agents Chemother.
Intern Med. 2010;49:1103-1109. 1996;40:1486-1490.
48. Van den Bulck K, Decostere A, Gruntar I, et al. In vitro antimicro- 64. Anacleto TP, Lopes LR, Andreollo NA, et al. Studies of distribution
bial susceptibility testing of Helicobacter felis, H. bizzozeronii, and and recurrence of Helicobacter spp. gastric mucosa of dogs after
H. salomonis. Antimicrob Agents Chemother. 2005;49:2997-3000. triple therapy. Acta Cir Bras. 2011;26:82-87.
49. Priestnall SL, Wiinberg B, Spohr A, et  al. Evaluation of 65. Velin D, Michetti P. Advances in vaccination against Helicobacter
­“Helicobacter heilmannii” subtypes in the gastric mucosas of cats pylori. Expert Rev Gastroenterol Hepatol. 2010;4:157-166.
and dogs. J Clin Microbiol. 2004;42:2144-2151. 66. Herrera V, Parsonnet J. Helicobacter pylori and gastric adenocarci-
50. Herbrink P, van Doorn LJ. Serological methods for diagnosis noma. Clin Microbiol Infect. 2009;15:971-976.
of Helicobacter pylori infection and monitoring of eradication 67. McColl KE. Clinical practice. Helicobacter pylori infection. N Engl
­therapy. Eur J Clin Microbiol Infect Dis. 2000;19:164-173. J Med. 2010;362:1597-1604.
51. Talebkhan Y, Ebrahimzadeh F, Esmaeili M, et  al. Helicobacter 68. Dieterich C, Wiesel P, Neiger R, et al. Presence of multiple “Heli-
pylori Omp18 and its application in serologic screening of ­infection. cobacter heilmannii” strains in an individual suffering from ulcers
Curr Microbiol. 2011;62:325-330. and in his two cats. J Clin Microbiol. 1998;36:1366-1370.
52. Seidel KE, Stolte M, Lehn N, et  al. Antibodies against Helico- 69. Lavelle JP, Landas S, Mitros FA, et  al. Acute gastritis associated
bacter felis in sera of cats and dogs. Zentralbl Veterinarmed B. with spiral organisms from cats. Dig Dis Sci. 1994;39:744-750.
1999;46:181-188. 70. Thomson MA, Storey P, Greer R, et  al. Canine-human transmis-
53. Strauss-Ayali D, Scanziani E, Deng D, et al. Helicobacter spp. infec- sion of Gastrospirillum hominis. Lancet. 1994;344:1097-1098.
tion in cats: evaluation of the humoral immune response and preva- 71. van Loon S, Bart A, den Hertog EJ, et al. Helicobacter heilmannii
lence of gastric Helicobacter spp. Vet Microbiol. 2001;79:253-265. gastritis caused by cat to child transmission. J Pediatr Gastroenterol
54. Strauss-Ayali D, Simpson KW, Schein AH, et  al. Serological dis- Nutr. 2003;36:407-409.
crimination of dogs infected with gastric Helicobacter spp. and 72. Meining A, Kroher G, Stolte M. Animal reservoirs in the transmis-
uninfected dogs. J Clin Microbiol. 1999;37:1280-1287. sion of Helicobacter heilmannii. Results of a questionnaire-based
55. Calvet X, Sanchez-Delgado J, Montserrat A, et  al. Accuracy of study. Scand J Gastroenterol. 1998;33:795-798.
diagnostic tests for Helicobacter pylori: a reappraisal. Clin Infect 73. De Groote D, Ducatelle R, Haesebrouck F. Helicobacters of
Dis. 2009;48:1385-1391. possible zoonotic origin: a review. Acta Gastroenterol Belg.
56. Cornetta AM, Simpson KW, Strauss-Ayali D, et al. Use of a [13C] 2000;63:380-387.
urea breath test for detection of gastric infection with Helicobacter 74. Minauchi K, Takahashi S, Sakai T, et  al. The nosocomial
spp in dogs. Am J Vet Res. 1998;59:1364-1369. ­transmission of Helicobacter cinaedi infections in immunocom-
57. Neiger R, Seiler G, Schmassmann A. Use of a urea breath test to promised patients. Intern Med. 2010;49:1733-1739.
evaluate short-term treatments for cats naturally infected with
Helicobacter heilmannii. Am J Vet Res. 1999;60:880-883.
CHAPTER 50

Leptospirosis
Jane E. Sykes

Recently, PCR-based DNA typing methods have been used


Overview of Leptospirosis to identify Leptospira strains on the basis of their genotype,
First Described: Heidelberg, Germany, 1886 (Adolf Weil)1 instead of using the serovar classification method. Specific geno-
Cause: Leptospira spp. (a spirochete) types appear to associate more strongly with particular reser-
voir hosts and disease manifestations than has been found for
Affected Hosts: Dogs, rarely cats, more than 150 other mam-
serovars.6,7 Multiple different leptospiral serovars have been
malian species
isolated from single outbreaks of human leptospirosis associ-
Geographic Distribution: Worldwide ated with specific clinical manifestations (such as pulmonary
Major Clinical Signs: Fever, lethargy, reluctance to move, hemorrhage).8 Therefore, virulence factors other than outer sur-
anorexia, polyuria and polydipsia, vomiting, diarrhea, face antigens (which define serovars) may more important deter-
icterus, respiratory difficulty minants of clinical manifestations of disease than the identity of
the infecting serovar.
Major Differential Diagnoses: Nephrotoxicoses (e.g., NSAIDs, Pathogenic Leptospira species do not replicate outside the host
grapes and raisins, ethylene glycol), Lyme nephritis, acute and are readily inactivated in the environment when exposed to
glomerulonephritides, bacterial pyelonephritis, acute excessive heat, ultraviolet irradiation, a variety of disinfectants,
pancreatitis, canine monocytic ehrlichiosis, Rocky Moun- and freezing conditions. However, when conditions are optimal,
tain Spotted Fever, bacterial sepsis, leishmaniosis pathogenic leptospires can survive in water and wet soil for weeks
Human Health Significance: Zoonosis. Human infection results to months.9 Dogs that drink from, swim, or wade in environmen-
from direct or indirect contact with contaminated urine. No tal water sources can become infected; exposure to wildlife or
signs, a mild influenza-like illness, or severe illness with mul- farm animals and their urine is also a risk factor. In developing
tiorgan failure (Weil’s disease) have the potential to occur. countries, dogs may be infected as a result of access to sewage.10
Although the “classic” signalment for a dog with leptospirosis is
a large-breed, outdoor, intact male dog, dogs of any age, breed,
and sex may be affected. For example, geriatric, small-breed dogs
Etiologic Agent and Epidemiology that live in urban or suburban areas and that contact rodents
or rodent urine can develop the disease. Indeed, over the past
Leptospirosis is a widespread zoonotic disease caused by systemic decade, residence in urban areas has emerged as a risk factor for
infection by pathogenic spirochetes of the genus Leptospira. Lep- Leptospira infection in dogs in some parts of North America,
tospira spp. are thin (0.1 µm in diameter), flexible, motile, spiral- possibly due to exposure to urban wildlife species.11 Cats may be
shaped bacteria that have a hook-shaped end (leptos = “thin,” infected after ingestion of infected prey, so outdoor exposure and
spira = “coiled”). A huge variety of different mammalian species hunting behavior may be risk factors for cats.2
may be infected by leptospires. These hosts can shed the organ- Outbreaks of disease in dogs have been correlated with periods
ism in their urine and contaminate the environment. Infection of of high annual rainfall and may occur approximately 3 months
incidental hosts results in disease that varies in severity from a after wet weather.12-14 The peak seasonal distribution in parts
mild febrile illness to severe multisystemic disease. Both dogs and of North America where freezing winters occur is late fall,11,15,16
cats can develop leptospirosis, but cats appear to be relatively but in more temperate regions, peak seasonal distribution occurs
resistant to development of clinical illness. As a result, leptospi- after months of high rainfall (such as in the winter in northern
rosis is uncommonly described in cats.2-5 California).13 Leptospirosis is most prevalent in parts of the world
Both pathogenic and saprophyte Leptospira species exist in where there is higher annual rainfall and a warm climate, but the
nature. Saprophytic species, such as Leptospira biflexa, live in presence of infected reservoir hosts also influences the geographic
water and soil and do not infect animal hosts. Disease in dogs distribution of leptospirosis. Within North America, dogs that live
is primarily caused by the pathogenic species Leptospira inter- in or travel to Hawaii, northern California, Oregon, Washington,
rogans and Leptospira kirschneri. Within the genus Leptospira, the upper Midwest and Midwest, parts of Texas, Colorado, and
more than 250 different serovars of pathogenic leptospires are the northeastern and mid-Atlantic coastal regions may be more
recognized, which are distinguished based on differences in their likely to develop infection.17 Leptospirosis also occurs in dogs in
lipopolysaccharide (LPS) O antigens. The serovars are grouped the southeastern United States,18 and the disease has been widely
into more than 20 antigenically related serogroups (Table 50-1). reported in dogs from Ontario in Canada.16 Numerous reports
At least 10 serovars have been associated with disease in dogs describe leptospirosis in dogs from across Europe.19 Major dis-
worldwide. Each serovar is adapted to one or more wild or ease foci among people include the Caribbean, Central and South
domestic animal reservoir host species (see Table 50-1). America, India, Southeast Asia, South America, and Oceania.

474
CHAPTER 50  Leptospirosis 475

TABLE 50-1
Selected Serovars of Leptospira interrogans and Leptospira kirschneri that Infect Dogs *
Species Serogroup Serovar Potential Reservoir Hosts Country
Leptospira Icterohaemorrhagiae Icterohaemorrhagiae Rats United States, France
interrogans Canicola Canicola Dogs India, United States
Pomona Pomona Horses, cattle, sea lions United States
Australis Bratislava Pigs, hedgehogs, horses United States
Sejroe ND Mice Germany
Ballum Ballum Mice, gray squirrels, muskrats, United States
opossums
Leptospira Grippotyphosa Grippotyphosa Raccoons, fox squirrels, muskrats, United States
kirschneri bobcats, voles, mice, rats, cattle,
horses

*Isolated from dogs with naturally-occurring leptospirosis, or that induce disease in dogs after experimental inoculation.

Worldwide, the most common serovars thought to infect


dogs before the introduction of canine Leptospira vaccines
several decades ago were Icterohaemorrhagiae and Canicola.20
Since that time, widespread seroconversion to other serovars,
especially Grippotyphosa and Pomona in North America, has
been noted in sick dogs. Increased testing for these serovars may
have contributed to this phenomenon, as well as increased con-
tact between dogs and wildlife and farm animal reservoir hosts
for these serovars.21 Serovars that currently cause disease in the
dog population are poorly understood, because the results of
serology do not accurately predict the infecting serovar,22 and
culture and identification of leptospiral serovars is insensitive,
requires significant technical expertise, and is not widely avail-
able (see Diagnosis, later).23

Clinical Features FIGURE 50-1  Uveitis in an Australian shepherd with leptospirosis with leakage of
serum bilirubin into the aqueous humor.
Signs and Their Pathogenesis
Pathogenic leptospires penetrate intact mucous membranes or show no signs of illness or develop only a mild, transient febrile
abraded skin. They multiply rapidly in the bloodstream as early illness. Severe clinical signs and death result from renal failure
as 1 day postinfection, which may be followed by the develop- and/or injury to the liver and lungs. Leptospirosis should be sus-
ment of vasculitis and multiorgan failure. The spirochetes can pected in dogs that develop signs of acute febrile illness, kidney
produce a variety of toxins, including phospholipases, sphingo- and hepatic injury, uveitis, or pulmonary hemorrhage; it should
myelinases, and pore-forming proteins. Infection follows direct also be suspected in breeding dogs with abortion, stillbirths, or
contact with infected urine or, more commonly, contact with neonatal deaths.20 Clinical signs in affected cats resemble those in
soil, water, food, or other fomites that have been contaminated dogs, although some cats have lacked evidence of hepatic injury.2
with infected urine. Transmission has also occurred as a result Fever occurs only at the onset of illness and is often accom-
of bite wound inoculation (such as from infected rodents), panied by shivering, reluctance to move, and generalized muscle
ingestion of infected tissues, and venereal and placental transfer. tenderness that may result from myositis. This may be followed
The incubation period for acute leptospirosis is approxi- by signs of lethargy, polyuria and polydipsia, inappetence, vom-
mately 7 days, but may be shorter or longer depending on the iting, and diarrhea.24-27 Gastrointestinal signs in some dogs may
dose and strain virulence of the leptospires involved, and the result from pancreatitis and enteritis, in addition to acute renal
host immune response.24 Dogs can also develop signs of chronic and liver damage. Polyuria and polydipsia may result from a
kidney disease months or more than a year after acute kidney decreased responsiveness of the inner medullary connecting ducts
injury. Early renal damage in this situation may be associated to antidiuretic hormone (acquired nephrogenic diabetes insipi-
with transient or undetected clinical signs, which are subse- dus). However, some dogs show decreased thirst and urination.
quently followed by either clearance or persistence of infection Liver involvement leads to icterus in some dogs (Figure 50-1).
and progression of renal damage. Longer incubation periods Components of leptospiral endotoxin such as oleic and linoleic
might occur in cats,2 but further study is required. acid are thought to interfere with hepatic transport pumps,
Leptospirosis varies considerably in severity, which may con- which leads to functional hepatic impairment.28 Hepatocyte
tribute to underrecognition of the disease. Many dogs probably apoptosis and injury to hepatocyte membranes is also suspected
476 SECTION 2  Bacterial Diseases

TABLE 50-2
Complete Blood Count Findings at Admission in Dogs with Leptospirosis from Northern California*
Percent Below Percent Within Percent Above Range for
Reference the Reference the Reference the Reference Dogs with Number
Test Range Range Range Range Leptospirosis Tested
Hematocrit (%) 40-55 78 22 0 12-48 54
Neutrophils (cells/µL) 3000-10,500 0 35 65 3474-14,311 54
Band neutrophils (cells/ Rare 0 63 37 0-2954 54
µL)
Monocytes (cells/µL) 150-1200 0 65 35 162-4594 54
Lymphocytes (cells/µL) 1000-4000 30 70 0 162-3769 54
Eosinophils (cells/µL) 0-1500 0 100 0 0-970 54
Platelets (cells/µL) 150,000- 28 61 11 40,000-522,000 53
400,000

*Diagnosis based on results of acute and convalescent phase serology together with consistent clinical signs.

to occur; leptospirosis in humans and guinea pig models is asso- strain. Severely affected dogs may show fever, dull mentation,
ciated with loss of the cellular adhesion molecule E-cadherin on dehydration, mild generalized peripheral lymphadenopathy,
hepatocytes.20 diffuse muscle pain, and pain on palpation of the abdomen
Less commonly, uveitis, myocardial damage, or reproductive or kidneys. Fever is typically only present early in the course
complications occur. Leptospiral DNA can be detected in the of infection. In oliguric or anuric dogs that have been treated
aqueous humor of human patients with leptospiral uveitis, and aggressively with intravenous fluids, evidence of overhydration
the development of uveitis may be delayed up to 18 months after may be present. Rarely, arrhythmias are detected on cardiac aus-
the onset of acute illness.29 ECG alterations can occur in dogs that cultation. Careful ophthalmologic examination may reveal con-
suggest myocardial damage.30 Abortion can occur during preg- junctivitis, scleral injection, and occasionally uveitis. Mild pallor,
nancy.31 Stillbirth and neonatal deaths have also been described. petechial or ecchymotic hemorrhages, and, less commonly, epi-
Bleeding tendencies in dogs with leptospirosis may be mani- staxis may be present. Respiratory difficulty, tachypnea, and
fested as petechial hemorrhages, hematuria, hematemesis, epi- harsh lung sounds may be evident in dogs that develop pulmo-
staxis, hematochezia, or melena.25,30,32 Although the precise nary complications. Dogs with liver involvement may be icteric.
mechanisms are unknown, bleeding tendencies may reflect the Rectal examination occasionally reveals melena or hematochezia.
presence of endothelial damage and disordered coagulation. In
human patients, thrombocytopenia (≤100,000 platelets/µL) and Diagnosis
prolonged PT have been associated with bleeding.33,34 Tachy-
pnea may be noted as a result of severe pulmonary hemorrhage Laboratory Abnormalities
syndrome (SPHS).35,36 SPHS is recognized commonly in humans Complete Blood Count
with severe leptospirosis, is not associated with significant lung The hemogram in dogs with leptospirosis may reveal neutro-
colonization by the spirochete, and seems to have an immune- philia, sometimes with bandemia; nonregenerative anemia;
mediated pathogenesis.37 The prevalence of SPHS in dogs appears lymphopenia; and/or thrombocytopenia (Table 50-2). Throm-
to be particularly high in continental Europe. For example, pul- bocytopenia occurs in up to 53% of dogs.25,26,30,41 The mecha-
monary complications occurred in more than two thirds of dogs nism is unclear; disseminated intravascular coagulation (DIC)
with leptospirosis in Berlin.36 Vasculitis can also contribute to the or immune-mediated mechanisms may be involved.
development of peripheral edema and mild pleural and perito-
neal effusions. In human patients, aseptic meningitis is a common Serum Biochemical Tests
complication of leptospirosis and results in signs of severe head- Findings on the serum chemistry panel vary depending on lep-
ache, delirium, and less commonly seizures and altered sensory tospiral strains that circulate in a particular geographic region
perception.38 Whether meningitis occurs in dogs is unknown. (Table 50-3). In all geographic locations, a combination of
The extent to which leptospires can cause chronic hepatic azotemia and elevated liver enzyme activities, especially when
injury in dogs is also unknown. In one report from France, lepto- thrombocytopenia is present, should raise suspicion for leptospi-
spires were detected in bile canaliculi of young dogs with chronic rosis. Azotemia is generally present in more than 80% to 90%
hepatitis using immunohistochemistry and electron microscopy. of dogs,25,26,30,42 although in one European study, increased
These dogs showed signs of poor growth, weight loss, and asci- serum creatinine concentration was present in only 57% of
tes.39,40 Attempts to detect Leptospira DNA in liver tissue from affected dogs.41 Uncommonly, dogs exhibit increased liver
dogs with chronic active hepatitis have been unrewarding. enzyme activities in the absence of azotemia. Signs of hepatic
dysfunction are associated with minimal histologic damage to
Physical Examination Findings the liver and include variable, and generally mild to moder-
Findings on physical examination of dogs with leptospirosis vary ate, increases in serum ALT, AST, and ALP activities and total
with disease severity, chronicity, and the infecting leptospiral bilirubin concentration.
CHAPTER 50  Leptospirosis 477

TABLE 50-3
Serum Biochemistry Findings at Admission in Dogs with Leptospirosis from Northern California*
Percent Below Percent Within Percent Above Range for
Test (Number of Reference the Reference the Reference the Reference Dogs with Number of
Dogs Tested) Range Range Range Range Leptospirosis Dogs Tested
Sodium (mmol/L) 145-154 33 61 6 134-162 54
Potassium (mmol/L) 4.1-5.3 35 44 20 2.8-7.6 54
Chloride (mmol/L) 105-116 57 37 6 98-118 54
Bicarbonate (mmol/L) 16-26 33 61 6 11-28 54
Calcium (mg/dL) 9.9-11.4 39 44 17 7.2-10.9 54
Phosphorus (mg/dL) 3.0-6.2 0 9 91 2.7-13.0 54
Creatinine (mg/dL) 0.5-1.6 0 0 100 0.4-3.3 54
BUN (mg/dL) 8-31 0 2 98 8-110 54
Albumin (g/dL) 2.9-4.2 96 4 0 0.9-3.8 54
Globulin (g/dL) 2.3-4.4 9 78 13 1.6-4.7 54
Cholesterol (mg/dL) 135-345 4 91 79-408 54
Total bilirubin (mg/ 0-0.4 0 69 32 0-3.1 54
dL)
ALT (U/L) 19-70 0 43 48 17-1443 54
ALP (U/L) 15-127 0 67 33 29-985 54
CK (U/L) 46-320 31 8 62 5-258,720 13

*Diagnosis based on results of acute and convalescent phase serology together with consistent clinical signs.

Other findings on the serum chemistry panel include fibrinogen, D-dimer, and fibrinogen degradation product con-
hypoalbuminemia and a variety of electrolyte abnormalities. centrations. Antithrombin activity may be decreased. Prolonga-
Hyponatremia, hypochloremia, or hyperphosphatemia occur tions in PT and PTT have been present in up to 15% of tested
in most dogs. Decreased expression of sodium transporters dogs.20,26,30 PT prolongations are associated with mortality in
by proximal convoluted tubule cells contributes to impaired human patients.34 In some dogs, a shortened PT is present,42
sodium resorption, increased distal sodium delivery, and potas- possibly because of DIC. In human patients, leptospirosis has
sium wasting.43 As a result, dogs with acute oliguric renal been associated with an imbalance between coagulation and
failure may have paradoxical hypokalemia. fibrinolysis.34
Moderately and occasionally markedly increased serum CK
activity is present in some dogs due to myositis. Increased serum Diagnostic Imaging
troponin concentrations have been found in some dogs with Plain Radiography
leptospirosis, which suggests myocardial damage.30 Increased Plain thoracic radiography in dogs with leptospirosis may show
serum pancreatic lipase immunoreactivity may occur, probably no abnormalities, or mild to severe, diffuse interstitial pulmo-
as a result of pancreatitis. nary patterns may be present. Thoracic radiographs from dogs
with SPHS can show severe nodular to diffuse interstitial and
Urinalysis alveolar patterns (Figure 50-2). Uncommonly, mild pleural
Findings on urinalysis in dogs with leptospirosis include isosthe- effusion is present.
nuria, occasionally hyposthenuria, and variable glucosuria, pro-
teinuria, pyuria, cylindruria, and bilirubinuria.20,26,30,42 Urine Sonographic Findings
protein to creatinine (UPC) ratios may be increased. Glucosuria Findings on abdominal sonography include mild renomegaly,
was present in 77% of 44 dogs with leptospirosis at the author’s increased renal cortical echogenicity, mild pyelectasia, perire-
institution, and in 21 dogs that had UPC ratios measured, the nal fluid accumulation or mild ascites, and a medullary band
ratios ranged from 0.4 to 11.6 (median, 3, reference range of increased echogenicity within the kidneys (Figure 50-3).45
less than 0.5). Most proteins in the urine are of low molecular None of these signs are specific for leptospirosis. Ultrasono-
weight, which supports a tubular rather than glomerular ori- graphic changes suggestive of pancreatitis are occasionally pres-
gin of these proteins.30,44 Leptospires are not visible in urine ent, such as enlargement and hypoechogenicity of the pancreas.
sediment by routine light microscopy. Thickening of the gastric, and less commonly small intestinal
wall may also be present. Mild to moderate hepatomegaly
Coagulation Function and splenomegaly with mottling of the splenic echotexture
Aside from thrombocytopenia, analysis of hemostatic func- may be detected, with or without evidence of mild abdominal
tion in dogs with leptospirosis shows variable increases in lymphadenomegaly.
478 SECTION 2  Bacterial Diseases

A
FIGURE 50-3  Abdominal sonogram image showing perirenal fluid accumulation in a
1.5-year-old male neutered kelpie with renal failure due to acute leptospirosis.

Microbiologic Tests
Diagnostic assays available for leptospirosis in dogs are
described in Table 50-4. Most often, diagnosis is based on acute
and convalescent phase serology.

Darkfield Microscopy
Examination of urine sediment using darkfield microscopy is
insensitive for diagnosis of leptospirosis. Considerable experi-
ence is required to accurately identify the spirochetes, so false
positives also have the potential to occur.

Culture
Culture of leptospires is not routinely performed, because it
B is slow, can be insensitive, and is not widely available. Special
leptospira growth media is required, such as Ellinghausen-
McCullough-Johnson-Harris (EMJH) medium. Cultures must
be incubated for up to 3 to 6 months, and overgrowth with
other bacteria can occur if cultures become contaminated.
Venous blood and/or urine should be collected using aseptic
technique before initiating antimicrobial drug treatment. Ide-
ally, a few drops of blood or urine are inoculated directly into
culture medium alongside the patient. In humans, blood is opti-
mally collected during the initial febrile period; urine can be
collected after the first week of illness. Because the exact time of
infection may be unknown, submission of both blood and urine
can increase the chance of a positive test result.
Only certain reference laboratories have expertise in culture
of leptospires and the ability to identify leptospires to the serovar
level, which is performed using a cross agglutinin absorption
test. Genetic typing methods such as pulsed field gel electropho-
C resis can also be performed.23 Despite the difficulties associated
with culture and identification of leptospires, a proper under-
FIGURE 50-2  Clinical images from a 4-year-old male neutered Havanese terrier with
standing of the epidemiology of leptospirosis depends on the
severe leptospiral pulmonary hemorrhage syndrome and anuric renal failure. A, Right lat-
eral thoracic radiograph. A severe, diffuse alveolar pattern is present. Mechanical ventila- use of these methods.
tion was needed and ultimately euthanasia was performed because of respiratory failure.
Histopathology of a lung biopsy showed severe pulmonary hemorrhage (B) and marked Serologic Diagnosis
alveolar histiocytosis (C). Currently the test of choice for diagnosis of leptospirosis is serol-
ogy using the microscopic agglutination test (MAT).20 In this
assay, the laboratory mixes serial dilutions of patient sera with
a panel of live leptospiral serovars. The laboratory then reports
the agglutinating antibody titer for each of the serovars tested,
CHAPTER 50  Leptospirosis 479

TABLE 50-4
Diagnostic Assays Available for Leptospirosis in Dogs
Assay Specimen Type Target Performance
Darkfield microscopy Urine Leptospira organisms Low sensitivity and specificity. Requires
considerable technical expertise to interpret
correctly.
Culture Whole blood, urine Leptospires Low sensitivity. Special media and prolonged
incubation times required. Difficult and not
widely available. Antimicrobial therapy may
lead to false-negative results. Currently the
only method that permits accurate serovar
identification.
Serology (MAT) Serum Antibodies against various False negatives can occur early in the course
leptospiral serovars of illness or with immunosuppression. False
positives can occur with a history of vac-
cination or with previous exposure. Paired
titers performed at the same laboratory
generally required for diagnosis. Interlabo-
ratory variation in results may occur.
Histopathology Kidney tissue collected Leptospires Organisms may be visualized with silver
via biopsy or stains, immunohistochemistry, or fluores-
necropsy cence in situ hybridization. Antimicrobial
therapy may lead to false-negative results.
PCR Blood, urine, tissue Leptospira DNA Sensitivity and specificity unclear and may
specimens vary between assays offered by different
laboratories. Antimicrobial therapy may
lead to negative PCR results.

which is the highest dilution of serum that causes the organisms levels (≥1600), especially if ongoing exposure to field strains
to agglutinate as assessed using darkfield microscopy. Seroposi- occurs. Thus, although single positive titers can raise suspicion
tive dogs can have high titers to multiple serovars tested because for the disease, they cannot be used to confirm the diagnosis
of serologic cross-reactivity. The highest titer does not necessar- of leptospirosis. After immunization, paradoxical cross-reac-
ily predict the infecting serovar.22 The serovar that produces the tivity to serogroups other than those used in the vaccine can
highest titer may also change when serology is repeated over the occur,46 so titers to non-vaccinal serovars in vaccinated dogs
course of illness.20 do not necessarily imply natural exposure to field strains.
Documentation of a fourfold or greater increase in titer to False-negative titers have the potential to occur if the
at least one serovar over time is usually required for diagnosis infecting serovar (or a related serovar) is not included in the
using the MAT, because titers are often low or negative in panel of serovars used to perform the test. Assays for diagno-
the first few days of illness, and a single high titer can result sis of human leptospirosis generally include a larger panel of
from previous exposure or a history of vaccination for lep- serovars (>20) than those used routinely in veterinary diagnos-
tospirosis. Later in the course of illness, a fourfold decrease tics (5 to 7 serovars). Assays should optimally include serovars
in titer may also be appropriate for diagnosis. Tradition- from serogroups that are known to circulate in the local dog
ally, convalescent titers are performed 2 to 4 weeks after the population, although this information is not often readily
acute titer, although seroconversion can sometimes occur as available.
early as 3 to 5 days after the initial titer is obtained. Nev- The MAT is widely available and relatively inexpensive,
ertheless, successive titers should be spaced at least 1 week but is complex to perform and interpret. Performance of the
apart, and it is critically important to use the same labora- test is hazardous to laboratory workers because live patho-
tory for each set of titers.20 Antimicrobial treatment has the genic serovars are used, and the test is difficult to standardize.
potential to blunt a rise in titer, but dogs usually serocon- Considerable interlaboratory variation in results can occur,
vert despite antimicrobial treatment.42 Titers that result from in terms of both the magnitude of the titers reported and the
previous vaccination, previous exposure or chronic infec- pattern of seroreactivity to each serovar included in the test.47
tion generally change by one dilution over time (e.g., from The identity of serovars used in the assay must be verified peri-
1:400 to 1:800) or remain static. Titers persist for at least odically to ensure that cross-contamination or culture dete-
1 year after natural infection, although they often decline by rioration has not occurred. A leptospirosis proficiency testing
4 months after vaccination.46 Nevertheless, postvaccinal titers scheme is offered by the International Leptospirosis Society
can persist for longer than a year and be maintained at high (ILS) to assist laboratories in the maintenance of quality
480 SECTION 2  Bacterial Diseases

assurance for the MAT.48,49 Veterinarians should strive to use


laboratories that participate in this quality assurance scheme.
Other serologic assays under investigation for diagnosis of
leptospirosis include ELISA assays for IgG and IgM antibod-
ies that are directed against leptospiral surface antigens.50-53
The performance of such assays have the potential to vary
geographically based on the serogroups that are prevalent in
the region. In Europe, a lateral-flow assay that detects IgM
(Test-it, Royal Tropical Institute/Life Assay Diagnostics) is
available for diagnosis of acute canine leptospirosis.50 This
assay appeared to have high clinical sensitivity for diagno-
sis of leptospirosis in dogs in the Netherlands (100%) and
the West Indies (78%), with a low rate of false positives
in healthy dogs from the Netherlands, the West Indies, and
Mexico (<5%).

Molecular Diagnosis Using the Polymerase Chain Reaction


Several veterinary diagnostic laboratories in the United States
and Europe offer PCR assays for the detection of the DNA of
pathogenic leptospires in blood and/or urine. The sensitivity
and specificity of these assays in dogs with leptospirosis has
not been thoroughly assessed and have the potential to vary
geographically, because of differences in shedding patterns
for different serovars. Assay performance also is likely to vary
from one laboratory to another. PCR assays have the poten-
tial to be advantageous for diagnosis early in the course of ill-
ness, when serologic tests are negative, or in dogs that possess
residual antibody after vaccination. Specimens for PCR testing
are best obtained before initiation of antimicrobial drug treat-
ment, and both blood and urine should be submitted in order
to optimize sensitivity. Negative results do not rule out a diag-
nosis of leptospirosis. Because subclinically infected dogs can FIGURE 50-4  Shrunken, irregular kidneys from a 16-year-old male neutered Jack
shed leptospires, a positive PCR test result on urine may not Russell terrier. The dog was treated for severe acute leptospirosis at 12 years of age. At
necessarily correlate with illness. Vaccination of healthy dogs that time, after six dialysis treatments, the dog was discharged from the hospital with a
with inactivated Leptospira vaccines should not lead to posi- creatinine of 4.2 mg/dL. Four years later, the dog died for reasons unrelated to renal injury
tive PCR assay results,54 so a history of Leptospira vaccination but had evidence of chronic renal injury at necropsy. (Image courtesy UC Davis Veterinary
should not interfere with PCR diagnosis. Currently available Anatomic Pathology Service.)
PCR assays do not provide information on the infecting serovar
or serogroup.
necrotizing myocarditis can also occur; myonecrosis is occa-
Pathologic Findings sionally described in skeletal muscle. Lung pathology in dogs
Gross Pathologic Findings with SPHS resembles that described in human patients and
Gross pathologic findings in dogs with leptospirosis include consists of intra-alveolar hemorrhage, pneumocyte necrosis,
jaundice, petechial and ecchymotic hemorrhages, and some- and hyaline membrane formation, in the absence of significant
times fibrin thrombi throughout the body. The lungs of dogs inflammation.
with SPHS may be wet, mottled, heavy, and dark-pink to red Silver stains such as Warthin-Starry stain and immunohisto-
in color.36 Ascites and/or pleural or pericardial effusion may chemistry can be used to visualize bacteria within tissue speci-
be present, and the intestinal lumen may be filled with blood mens (see Figure 50-5, B). Fluorescence in situ hybridization
or contain melena. The kidneys of dogs with acute disease may has also been used to identify and localize leptospires within the
be mildly enlarged, pale and sometimes mottled or petechiated; kidneys (see Figure 5-1).
they may also contain infarcts. Dogs with chronic renal injury
secondary to the infection can have kidneys that are shrunken Treatment and Prognosis
and irregular (Figure 50-4).
Antimicrobial Drugs
Histopathologic Findings Penicillins or doxycycline are recommended for initial treat-
Infection of the kidneys by leptospires leads to an interstitial ment of humans and dogs with leptospirosis. Treatment
nephritis, with mixed inflammatory infiltrates. Acute tubular should be initiated as early as possible, before the results of
necrosis can also occur as a result of vasculitis and renal isch- diagnostic assays become available. In hamster models, doxy-
emia (Figure 50-5, A).36 Histopathologic changes in the liver cycline clears spirochetes from all tissues, including the kidney,
are often mild and include mild random hepatic necrosis, mild within 3 days of infection; ampicillin is less effective at clear-
neutrophilic periportal hepatitis, cholestasis, or liver plate dis- ing organisms from the kidney.55 Based on current evidence,
array (Figure 50-6). Multifocal, random, neutrophilic, and the ACVIM Consensus Statement on leptospirosis in dogs
CHAPTER 50  Leptospirosis 481

B
FIGURE 50-6  Histopathology of the liver in leptospirosis. A, Minimal hepatic changes
and severe cholestasis in an 8-year-old male neutered Australian shepherd mix that was
euthanized because of anuric leptospirosis. The activity of serum ALP and the serum total
B bilirubin concentration were mildly increased, and the dog seroconverted with the highest
titer to serovar Pomona (1:400 to >1:3200 12 days later). B, Liver plate disarray with hepa-
FIGURE 50-5  A, Histopathology of the kidney of a dog that died of leptospirosis. tocellular dissociation in a dog with leptospirosis. (B, Image courtesy John Cullen, North
Acute tubular necrosis and interstitial nephritis is present. H&E stain. B, Warthin-Starry Carolina State University and Jean-Jacques Fontaine, École Nationale Vétérinaire d’Alfort)
showing low numbers of leptospires in the renal interstitium (arrow). (Images courtesy Dr.
Patricia Pesavento, University of California, Davis Veterinary Anatomic Pathology Service.)

TABLE 50-5
Antimicrobials Recommended for Treatment of Leptospirosis
recommends that all dogs with leptospirosis should be treated in Dogs
with doxycycline, 5 mg/kg q12h PO or IV for 2 weeks.20 If
vomiting or other adverse reactions preclude the use of doxy- Interval Duration
cycline, parenteral ampicillin or penicillin G can be used (Table Drug Dose Route (hours) (days)
50-5). Dogs initially treated with a penicillin derivative should
be treated with doxycycline for 2 weeks after gastrointestinal Doxycycline 5 mg/kg PO, IV 12 14
signs resolve. Fluoroquinolones are not highly efficacious for Ampicillin 20 mg/kg* IV 6 Variable
clearance of leptospires from tissues.56 Other antimicrobials Penicillin 25,000- IV 12 Variable
that have been efficacious in clinical trials involving human 40,000 U/kg
patients with leptospirosis include ceftriaxone, cefotaxime,
and azithromycin.57,58 *Reduce dose in renal failure.
482 SECTION 2  Bacterial Diseases

Supportive Care natural infection is unknown. Immunity after vaccination with


Dogs with severe disease require aggressive treatment with IV inactivated vaccines is serogroup specific and possibly serovar
crystalloids, potassium supplementation, antiemetics, antihy- specific, although partial immunity to heterologous serogroups
pertensives, antacids, blood products, and parenteral or enteral was shown in one report.61 A live attenuated LPS mutant L.
nutrition, with attention to precautions to minimize zoonotic interrogans serovar Manilae vaccine was shown to induce
transmission (see Public Health Aspects, later). Referral to a strong protection in hamsters against challenge either with the
24-hour care facility, ideally with access to dialysis, should be same or a serologically unrelated (Pomona) serovar.62 After
considered for severely ill dogs because of the need for intensive challenge with serovar Pomona, survival rates were 100% in
monitoring and close observation. Electrolyte and acid-base sta- hamsters immunized with the attenuated live vaccine, com-
tus may require frequent monitoring. pared with only 40% for those immunized with an inactivated
Fluid therapy is critical and should be calculated based on serovar Manilae vaccine.
measurement of “outs and ins,” together with careful attention Current vaccines for dogs are inactivated bacterins. Bivalent
to body weight, respiratory rate, lung sounds, blood pressure, vaccines that contained only serovars Icterohaemorrhagiae and
and if possible, central venous pressure. Dogs with polyuric Canicola were first introduced for prevention of leptospirosis
renal failure may require extremely high fluid rates. For dogs in dogs. In the United States, these have now been replaced
with oliguric or anuric renal failure, high fluid rates can lead with four-serovar vaccines that contain serovars Icterohaem-
to overhydration and respiratory failure. Urine output in dogs orrhagiae, Canicola, Grippotyphosa, and Pomona. In Europe,
with oliguria or anuria should be closely monitored through serovar Icterohaemorrhagiae and Canicola vaccines are still
use of a closed, indwelling urinary catheter and collection bag available, but vaccines that contain serovar Icterohaemor-
system. Diuretics such as furosemide and mannitol may be indi- rhagiae, Canicola, and Grippotyphosa (Pfizer Animal Health)
cated. Failure of urine production after adequate hydration is and serovar Copenhageni (serogroup Icterohaemorrhagiae),
achieved is an indication for dialysis (see Renal Replacement Portland-vere (serogroup Canicola), Bratislava (serogroup Aus-
Therapy, next). Once recovery of kidney values has occurred, tralis), and Dadas (serogroup Grippotyphosa) (MSD Animal
fluids should be tapered slowly before they are discontinued to Health) have been introduced. Leptospiral vaccines effectively
ensure that proper hydration status is maintained. In some dogs, prevent disease and reduce shedding after challenge with the
possibly because of enteritis or pancreatitis, inappetence persists serovar included in the vaccine. The duration of immunity fol-
for several days after kidney and liver values normalize. Contin- lowing vaccination is at least 12 months.32,63,64 Leptospirosis
ued nutritional support is required during this period. has occurred in North American dogs immunized with bivalent
Dogs with SPHS may require oxygen therapy and, rarely, vaccines; however, disease is rare in dogs immunized with four-
mechanical ventilation. The optimum treatment for dogs serovar vaccines.13
with SPHS is unknown. Human patients with SPHS have had In the past, vaccination for with Leptospira vaccines has
improved outcome following cyclophosphamide therapy and been associated with type I hypersensitivity reactions such as
plasma exchange.59 anaphylaxis, especially in small-breed dogs. Anecdotal evidence
from industry and veterinary practitioners in North America
Renal Replacement Therapy suggests that the prevalence of these reactions has considerably
Early hemodialysis results in improved survival and shorter hos- reduced in recent years (to <1%) following efforts from industry
pital stays in human patients with leptospirosis.60 Renal replace- to remove constituents that have been associated with vaccine
ment therapy is indicated for dogs with inadequate urine output reactivity.20
that develop volume overload, hyperkalemia, a BUN greater than Immunization is recommended for dogs at risk of expo-
80 mg/dL, or signs of uremia that do not respond to medical man- sure. In some regions, this may be dogs that contact wild-
agement.20 Approximately 50% of 89 dogs with leptospirosis at life or farm animals, or dogs that swim, wade, or drink from
the University of California, Davis, VMTH in the past 10 years environmental water sources. In other regions, dogs in urban
received hemodialysis; the median number of hemodialysis treat- or suburban backyards may be at risk if there is significant
ments required was 3 (range, 1 to 14). Recovery of adequate renal exposure to wildlife or rodents in the immediate home envi-
function usually occurs within 2 to 4 weeks of starting dialysis. ronment. Immunization may be optimally performed a few
months before the onset of the peak season for leptospirosis
Prognosis in a given geographic region. There is no evidence that the
When the treatment is initiated early and aggressively, the prog- initial series needs to be readministered to dogs that are over-
nosis for recovery from acute leptospirosis is excellent. Survival due for an annual booster. For dogs that experience natural
rates are in excess of 50%,41 and in centers where hemodialysis is infection, immunization could commence a year after recov-
available, survival rates can exceed 80%. Precise survival rates for ery, although the duration of immunity after natural infection
dogs with SPHS are unknown but may be lower. Successful treat- and the degree and duration of cross-protection to heterolo-
ment is associated with normalization of the platelet count, serum gous serovars that follows natural infection requires further
BUN and creatinine concentrations, and serum liver enzyme activ- investigation.
ities within 10 to 14 days. Icterus may be slow to resolve, and
urine-concentrating ability may not return for at least 4 weeks. Prevention
Some dogs have residual permanent renal damage after recovery.
Leptospirosis can be prevented by restricting dogs’ access to
Immunity and Vaccination potential reservoir host species or environmental water sources.
Access to wildlife and farm animals can be reduced through
Recurrence of leptospirosis in dogs after recovery from natural fencing and rodent control. Where this is not possible, immuni-
infection appears to be rare, but the duration of immunity after zation is recommended.
CHAPTER 50  Leptospirosis 483

Public Health Aspects


BOX 50-1
In humans, leptospirosis occurs after an incubation period of 2
to 20 days (mean, 10 days). In 90% of infections, it is a mild,
Suggested In-Hospital Precautions for Dogs with Known
influenza-like illness. Less commonly, it is manifested by severe or Suspected Leptospirosis
multiorgan failure, with renal failure, hepatic failure, or SPHS.
Abortion can occur during pregnancy. Weil’s disease, which Warning signage on the cage or run
is characterized by impaired renal and hepatic function, usu- Minimize movement around the hospital, avoiding trans-
ally occurs a week after an initial illness characterized by fever, port along common hallways
myalgia, headache, chills, and conjunctivitis.1 Gloves and disposable gowns
In developed countries, most human leptospirosis cases fol- Protective eyewear and face shields if aerosolization of
low recreational activities that involve water, such as house- urine likely
boating or participation in triathlons, caving, or white-water Handwashing before and after handling
rafting.65,66 Individuals who contact farm animals can also No pregnant or immunocompromised staff to work with
develop leptospirosis. Contact with wild rodents adopted as pets the patient
has also led to human disease.67 In developing countries, stray Prompt disinfection of urine and urine spills
dogs may be a source of human infection, although rodents are Treatment with appropriate antimicrobial drugs
also important in this situation.68 The extent to which domestic Indwelling rather than intermittent urinary catheterization
cats might serve as reservoir hosts is not understood. for monitoring urine output
Reports of transmission from clinically affected incidental Dogs walked or gurneyed outside to urinate frequently if
hosts (as opposed to subclinically infected reservoir hosts) to urinary catheter not in place
other animals are rare, and there are no reports that document Urination outside in a designated area that can be easily
transmission of leptospires from pet dogs to humans with the disinfected
support of molecular typing methods. Appropriate antimicro- Warnings to others who handle the dog or specimens from
bial therapy may also reduce the possibility of transmission to the dog
humans. Nevertheless, precautions should be taken in the hospi-
tal environment to minimize zoonotic transmission (see Chapter
11). All dogs with acute renal failure should be handled as lep-
tospirosis suspects until an alternative diagnosis is established. and gloved hands should be used to clean up urinary accidents
Suggested precautions are shown in Box 50-1. Urine from dogs that occur indoors. Dogs should be taken to urinate in a place
with leptospirosis can be inactivated with a variety of disin- that is away from water and away from other pets and peo-
fectant solutions, including iodine-based disinfectants, quater- ple, especially children. Owners should wash their hands after
nary ammonium solutions, and accelerated hydrogen peroxide. they handle their pets. Veterinarians should recommend that
Large volumes of urine can be inactivated by 1:1 dilution with owners seek medical attention if illness occurs around the time
a 10% solution of household bleach. In dogs with indwelling their dog is diagnosed with leptospirosis, or if they have any
urinary catheters, 10% bleach should be injected directly into questions about the disease in humans. Immunocompromised
the collection bag before disposal. All blood, urine, and tissues humans should be referred to their medical practitioners for
from dogs should be treated as medical waste. If a dog dies or advice. Owners should also be informed that environmental
is euthanized, individuals who are to handle the remains should sources of infection may represent an ongoing risk, not only to
be alerted of the possibility of leptospirosis. other dogs in the household but also to humans. If other dogs
Owners of affected dogs should be educated in regard to the in the household may have been exposed at the same time as the
zoonotic nature of the disease. The risk of infection to owners dog diagnosed with leptospirosis, those dogs should be treated
is probably low, because urinary shedding generally does not with doxycycline for 2 weeks, because subclinical infection and
occur until 7 to 10 days after the onset of illness, and doxy- shedding may have occurred.20
cycline treatment likely results in clearance of bacteria from Veterinarians should contact their local or state health
the kidneys and urine within 2 to 3 days. Nevertheless, until department or the Centers for Disease Control and Prevention
antimicrobial therapy is completed, owners should avoid con- if additional questions arise regarding the public health risks
tact with their dog’s urine. Routine household disinfectants and zoonotic transmission of leptospirosis.20
484 SECTION 2  Bacterial Diseases

CASE EXAMPLE WBC 8820 cells/µL (6000-13,000 cells/µL)


Neutrophils 7673 cells/µL (3000-10,500 cells/µL)
Lymphocytes 353 cells/µL (1000-4000 cells/µL)
Monocytes 441 cells/µL (150-1200 cells/µL)
Platelets 74,000 platelets/µL (150,000-400,000 platelets/µL).
Serum Chemistry Profile:
Sodium 143 mmol/L (145-154 mmol/L)
Potassium 3.7 mmol/L (4.1-5.3 mmol/L)
Chloride 104 mmol/L (105-116 mmol/L)
Bicarbonate 17 mmol/L (16-26 mmol/L)
Phosphorus 5.1 mg/dL (3.0-6.2 mg/dL)
Calcium 9.6 mg/dL (9.9-11.4 mg/dl)
BUN 90 mg/dL (8-31 mg/dL)
Creatinine 4.6 mg/dL (0.5-1.6 mg/dL)
Glucose 110 mg/dL (70-118 mg/dL)
Total protein 5.1 g/dL (5.4-7.4 g/dL)
Albumin 2.4 g/dL (2.9-4.2 g/dL)
Globulin 2.7 g/dL (2.3-4.4 g/dL)
ALT 42 U/L (19-67 U/L)
FIGURE 50-7  “Makalu,” a 1.5-year-old male neutered kelpie dog that developed AST 146 U/L (21-54 U/L)
leptospirosis. ALP 73 U/L (15-127 U/L)
Creatine kinase 1387 U/L (46-320 U/L)
Signalment: “Makalu,” a 1.5-year-old male neutered kelpie GGT 2 U/L (0-6 U/L)
dog from Dixon in northern California (Figure 50-7). Cholesterol 189 mg/dL (135-345 mg/dL)
History: Makalu had a 1-day history of lethargy and inappetence. Total bilirubin 0.4 mg/dL (0-0.4 mg/dL).
The owner, a veterinarian, took his temperature at home and Urinalysis: SGr 1.013; pH 7.5, 2+ protein (SSA), 1+ bilirubin, 3+
it was >104°F (40°C). There had been no vomiting, diarrhea, hemoprotein, 2+ glucose, 0 WBC/HPF, 20-30 RBC/HPF, rare
coughing, sneezing, or increased thirst and urination. Makalu transitional epithelial cells.
lived on a property with another dog, three cats, sheep, goats, Coagulation Profile: PT 8.8 (7.5-10.5), APTT 16.8 (9-12),
and a pot-bellied pig. He normally ate a commercial dry dog fibrinogen 280 (90-255).
food; he consumed a rawhide the day before he became Serum Ethylene Glycol Assay: Negative.
ill. There was no history of previous travel, toxins, trauma, Imaging Findings:
medications, or tick exposure. He was 3 months overdue for Thoracic Radiographs: The cardiovascular and pulmonary
vaccination but had been vaccinated as a puppy for distemper, structures appeared within normal limits.
hepatitis, parvovirus, rabies, and leptospirosis. The owner did Abdominal Ultrasound: The liver and spleen were both
not recall which Leptospira vaccines he received. enlarged. The tail of the spleen had a mildly mottled
Physical Examination: echotexture. Both kidneys were mildly enlarged, and
Body Weight: 22.4 kg measured approximately 8 cm in length. The renal cortices
General: Quiet, alert and responsive. Ambulatory on all four limbs, were mildly echogenic. A small amount of subcapsular and
T = 103.7°F (39.8°C), HR = 80 beats/min, RR = 20 breaths/min, retroperitoneal fluid was noted in association with both
mucous membranes pink, CRT = 1 s. Esti­mated to be 7% to 8% kidneys. The mesenteric lymph nodes were prominent.
dehydrated based on skin turgor. Microbiologic Testing: Aerobic bacterial culture of blood
Integument, Eyes, Ears, Nose, and Throat: Mild scleral and urine: No growth.
injection and chemosis were present bilaterally. Leptospira serology (MAT): Day 2 after the onset of illness, nega-
Musculoskeletal: Appeared stiff, but there was no evidence of tive for antibodies to serovars Canicola, Grippotyphosa,
synovial effusion. Vocalized on flexion of the right thoracic Hardjo, Icterohaemorrhagiae, and Pomona at 1:100. Posi-
limb. tive for antibodies to serovar Bratislava at 1:100. Day 10 af-
Cardiovascular: Strong femoral pulses. No murmurs or ter the onset of illness, negative for antibodies to serovar
arrhythmias auscultated. Systolic blood pressure was Canicola. Positive for antibodies to serovars Grippotyphosa
145 mm Hg. (1:400), Hardjo (1:100), Icterohaemorrhagiae (1:800), Pomo-
Respiratory: Quiet bronchovesicular sounds were present in na (1:12,800), and Bratislava (1:3200).
all lung fields. Diagnosis: Leptospirosis.
Gastrointestinal and Genitourinary: A tense abdomen was Treatment: Makalu was aggressively treated with intra­venous
noted, with abdominal splinting noted on palpation. No fluids, mannitol, famotidine, a metoclopramide, and ampicillin
abnormalities were detected on rectal examination. (20 mg/kg IV q6h). Despite this treatment and administration
Lymph Nodes: No abnormalities were detected. of furosemide, urine output remained at <0.8 mL/kg/hr. After
Laboratory Findings: 18 hours of hospitalization, dialysis was initiated. After a second
CBC: dialysis treatment 2 days later, polyuria ensued (Table 50-6). On
HCT 41.4% (40%-55%) day 3, DIC developed, with further prolongation of the PT (14.3
MCV 70.2 fL (65-75 fL) s) and APTT (21.1 s), hypofibrinogenemia (<90 mg/dL), and
MCHC 36.0 g/dL (33-36 g/dL) a positive D-dimer assay. The dog was treated with multiple
CHAPTER 50  Leptospirosis 485

units of fresh frozen plasma. Treatment with ondansetron was antibody production to occur. The diagnosis of leptospirosis
also initiated on day 3 because of persistent vomiting, and was based on consistent clinical signs together with
total parenteral nutrition was instituted on day 4. Makalu’s seroconversion on day 10. Liver enzyme activities were
mentation improved on day 5 and he began eating on day 8. not increased in this dog on admission, but increased
Comments: Antibody titers in this dog were very low or  nega­ subsequently. Without dialysis, this dog would have died
tive initially because insufficient time had elapsed for from the disease.

TABLE 50-6
Serial Changes in Clinicopathologic Variables in a Hospitalized Dog with Leptospirosis That Was Treated with Hemodialysis
Variable Day 1 Day 3 Day 4 Day 5 Day 8 Day 10
ALT (U/L) 62 ND 105 110 82 71
ALP (U/L) 83 ND 97 97 79 75
Bilirubin (mg/dL) 0.4 ND 0.5 0.8 0.3 0.3
BUN (mg/dL) 105 144 54 47 35 27
(27)* (30)
Creatinine (mg/dL) 5.6 12.9 4.8 3.3 2.1 1.6
(2.2) (3.1)
Albumin (g/dL) 2.2 2.4 2.5 2.9 3.0 3.1
Platelets (x1000/µL) 74 ND 26 91 92 clumped
Urine output (mL/kg/hr) <0.8 0.25 10 10 7 ND

*Values in parentheses are postdialysis values. ND, not done.

SUGGESTED READINGS 12. Adin CA, Cowgill LD. Treatment and outcome of dogs with leptospi-
rosis: 36 cases (1990-1998). J Am Vet Med Assoc. 2000;216:371-375.
Araujo ER, Seguro AC, Spichler A, et al. Acute kidney injury in human 13. Hennebelle J, Sykes JE, Carpenter T, et  al. Spatial and temporal
leptospirosis: an immunohistochemical study with pathophysiologi- patterns of Leptospira spp. seroreactivity in dogs from northern
cal correlation. Virchows Arch. 2010;456(4):367-375. California. J Am Vet Med Assoc. 2012.
Sykes JE, Hartmann K, Lunn KF, et al. 2010 ACVIM small animal con- 14. Ward MP. Seasonality of canine leptospirosis in the United States
sensus statement on leptospirosis: diagnosis, epidemiology, treatment and Canada and its association with rainfall. Prev Vet Med.
and prevention. J Vet Intern Med. 2011;25(1):1-13. 2002;56:203-213.
15. Gautam R, Guptill LF, Wu CC, et al. Spatial and spatio-­temporal
REFERENCES clustering of overall and serovar-specific Leptospira microscopic
1. Levett PN. Leptospirosis. Clin Microbiol Rev. 2001;14:296-326. agglutination test (MAT) seropositivity among dogs in the United
2. Arbour J, Blais M, Carioto L, et al. Clinical leptospirosis in three States from 2000 through 2007. Prev Vet Med. 2010;96:122-131.
cats (2001-2009). J Am Anim Hosp Assoc. 2012;48:256-260. 16. Prescott J. Canine leptospirosis in Canada: a veterinarian’s perspec-
3. Bryson DG, Ellis WA. Leptospirosis in a British domestic cat. tive. CMAJ. 2008;178:397-398.
J Small Anim Pract. 1976;17:459-465. 17. Moore GE, Guptill LF, Glickman NW, et al. Canine leptospirosis,
4. Mason RW, King SJ, McLachlan NM. Suspected leptospirosis in United States, 2002-2004. Emerg Infect Dis. 2006;12:501-503.
two cats. Aust Vet J. 1972;48:622-623. 18. Brown CA, Roberts AW, Miller MA, et al. Leptospira interrogans
5. Rees HG. Leptospirosis in a cat. New Zealand Vet J. 1964;12:64. serovar grippotyphosa infection in dogs. J Am Vet Med Assoc.
6. Zuerner RL, Alt DP. Variable nucleotide tandem-repeat analy- 1996;209:1265-1267.
sis revealing a unique group of Leptospira interrogans serovar 19. Ellis WA. Control of canine leptospirosis in Europe: time for a
Pomona isolates associated with California sea lions. J Clin Micro- change? Vet Rec. 2010;167:602-605.
biol. 2009;47:1202-1205. 20. Sykes JE, Hartmann K, Lunn KF, et  al. 2010 ACVIM small ani-
7. Timoney JF, Kalimuthusamy N, Velineni S, et al. A unique genotype mal consensus statement on leptospirosis: diagnosis, epidemiology,
of Leptospira interrogans serovar Pomona type kennewicki is asso- treatment, and prevention. J Vet Intern Med. 2011;25:1-13.
ciated with equine abortion. Vet Microbiol. 2011;150:349-353. 21. Rentko VT, Clark N, Ross LA, et al. Canine leptospirosis. A retro-
8. Trevejo RT, Rigau-Perez JG, Ashford DA, et al. Epidemic leptospi- spective study of 17 cases. J Vet Intern Med. 1992;6:235-244.
rosis associated with pulmonary hemorrhage—Nicaragua, 1995. 22. Levett PN. Usefulness of serologic analysis as a predictor of the
J Infect Dis. 1998;178:1457-1463. infecting serovar in patients with severe leptospirosis. Clin Infect
9. Zaitsev SV, Chernukha Iu G, Evdokimova OA, et al. Survival rate Dis. 2003;36:447-452.
of Leptospira pomona in the soil at a natural leptospirosis focus. 23. Galloway RL, Levett PN. Application and validation of PFGE for
Zh Mikrobiol Epidemiol Immunobiol. 1989:64-68. serovar identification of Leptospira clinical isolates. PLoS Negl
10. Meeyam T, Tablerk P, Petchanok B, et al. Seroprevalence and risk Trop Dis. 2010;4:e824.
factors associated with leptospirosis in dogs. Southeast Asian J 24. Greenlee JJ, Alt DP, Bolin CA, et  al. Experimental canine lepto-
Trop Med Public Health. 2006;37:148-153. spirosis caused by Leptospira interrogans serovars pomona and
11. Raghavan R, Brenner K, Higgins J, et al. Evaluations of land cover bratislava. Am J Vet Res. 2005;66:1816-1822.
risk factors for canine leptospirosis: 94 cases (2002-2009). Prev Vet
Med. 2011;101:241-249.
486 SECTION 2  Bacterial Diseases

25. Birnbaum N, Barr SC, Center SA, et  al. Naturally acquired lep- 48. Chappel RJ, Goris M, Palmer MF, et al. Impact of proficiency test-
tospirosis in 36 dogs: serological and clinicopathological features. ing on results of the microscopic agglutination test for diagnosis of
J Small Anim Pract. 1998;39:231-236. leptospirosis. J Clin Microbiol. 2004;42:5484-5488.
26. Goldstein RE, Lin RC, Langston CE, et  al. Influence of infecting 49. International Leptospirosis MAT Proficiency Testing Scheme. 2013.
serogroup on clinical features of leptospirosis in dogs. J Vet Intern http://www.med.monash.edu.au/microbiology/staff/adler/proftemp.
Med. 2006;20:489-494. html. Last accessed February 3, 2013.
27. Greenlee JJ, Bolin CA, Alt DP, et al. Clinical and pathologic compari- 50. Abdoel TH, Houwers DJ, van Dongen AM, et  al. Rapid test for
son of acute leptospirosis in dogs caused by two strains of Leptospira the serodiagnosis of acute canine leptospirosis. Vet Microbiol.
kirschneri serovar grippotyphosa. Am J Vet Res. 2004;65:1100-1107. 2011;150:211-213.
28. Burth P, Younes-Ibrahim M, Santos MC, et al. Role of nonesteri- 51. Chalayon P, Chanket P, Boonchawalit T, et al. Leptospirosis sero-
fied unsaturated fatty acids in the pathophysiological processes of diagnosis by ELISA based on recombinant outer membrane pro-
leptospiral infection. J Infect Dis. 2005;191:51-57. tein. Trans R Soc Trop Med Hyg. 2011;105:289-297.
29. Chu KM, Rathinam R, Namperumalsamy P, et al. Identification of Lep- 52. La-Ard A, Amavisit P, Sukpuaram T, et al. Evaluation of recom-
tospira species in the pathogenesis of uveitis and determination of clinical binant Lig antigen-based ELISA for detection of leptospiral anti-
ocular characteristics in south India. J Infect Dis. 1998;177:1314-1321. bodies in canine sera. Southeast Asian J Trop Med Public Health.
30. Mastrorilli C, Dondi F, Agnoli C, et  al. Clinicopathologic fea- 2011;42:128-137.
tures and outcome predictors of Leptospira interrogans Austra- 53. Sun A, Wang Y, Du P, et  al. A sensitive and specific IgM-ELISA
lis ­serogroup infection in dogs: a retrospective study of 20 cases for the serological diagnosis of human leptospirosis using a
(2001-2004). J Vet Intern Med. 2007;21:3-10. rLipL32/1-LipL21-OmpL1/2 fusion protein. Biomed Environ Sci.
31. Rossetti CA, Liem M, Samartino LE, et  al. Buenos Aires, a new 2011;24:291-299.
Leptospira serovar of serogroup Djasiman, isolated from an 54. Midence JN, Leutenegger CM, Chandler AM, et al. Effects of recent
aborted dog fetus in Argentina. Vet Microbiol. 2005;107:241-248. Leptospira vaccination on whole blood real-time PCR testing in
32. Minke JM, Bey R, Tronel JP, et al. Onset and duration of protective healthy client-owned dogs. J Vet Intern Med. 2011;26:149-152.
immunity against clinical disease and renal carriage in dogs pro- 55. Truccolo J, Charavay F, Merien F, et al. Quantitative PCR assay
vided by a bi-valent inactivated leptospirosis vaccine. Vet Micro- to evaluate ampicillin, ofloxacin, and doxycycline for treatment
biol. 2009;137:137-145. of experimental leptospirosis. Antimicrob Agents Chemother.
33. Chierakul W, Tientadakul P, Suputtamongkol Y, et al. Activation 2002;46:848-853.
of the coagulation cascade in patients with leptospirosis. Clin Infect 56. Griffith ME, Moon JE, Johnson EN, et al. Efficacy of fluoroquino-
Dis. 2008;46:254-260. lones against Leptospira interrogans in a hamster model. Antimi-
34. Wagenaar JF, Goris MG, Partiningrum DL, et al. Coagulation disor- crob Agents Chemother. 2007;51:2615-2617.
ders in patients with severe leptospirosis are associated with severe 57. Panaphut T, Domrongkitchaiporn S, Vibhagool A, et  al. Ceftri-
bleeding and mortality. Trop Med Int Health. 2010;15:152-159. axone compared with sodium penicillin G for treatment of severe
35. Kohn B, Steinicke K, Arndt G, et al. Pulmonary abnormalities in leptospirosis. Clin Infect Dis. 2003;36:1507-1513.
dogs with leptospirosis. J Vet Intern Med. 2010;24:1277-1282. 58. Phimda K, Hoontrakul S, Suttinont C, et  al. Doxycycline versus
36. Klopfleisch R, Kohn B, Plog S, et al. An emerging pulmonary haem- azithromycin for treatment of leptospirosis and scrub typhus. Anti-
orrhagic syndrome in dogs: similar to the human leptospiral pulmo- microb Agents Chemother. 2007;51:3259-3263.
nary haemorrhagic syndrome? Vet Med Int. 2010;2010:928541. 59. Trivedi SV, Vasava AH, Bhatia LC, et  al. Plasma exchange with
37. Croda J, Neto AN, Brasil RA, et al. Leptospirosis pulmonary haem- immunosuppression in pulmonary alveolar haemorrhage due to
orrhage syndrome is associated with linear deposition of immuno- leptospirosis. Indian J Med Res. 2010;131:429-433.
globulin and complement on the alveolar surface. Clin Microbiol 60. Cerqueira TB, Athanazio DA, Spichler AS, et al. Renal involvement
Infect. 2010;16:593-599. in leptospirosis—new insights into pathophysiology and treatment. Braz
38. Jha S, Ansari MK. Leptospirosis presenting as acute meningoen- J Infect Dis. 2008;12:248-252.
cephalitis. J Infect Dev Ctries. 2010;4:179-182. 61. Sonrier C, Branger C, Michel V, et  al. Evidence of cross-protec-
39. Adamus C, Buggin-Daubie M, Izembart A, et al. Chronic hepatitis tion within Leptospira interrogans in an experimental model. Vaccine.
associated with leptospiral infection in vaccinated beagles. J Comp 2000;19:86-94.
Pathol. 1997;117:311-328. 62. Srikram A, Zhang K, Bartpho T, et al. Cross-protective immunity
40. Bishop L, Strandberg JD, Adams RJ, et al. Chronic active hepatitis in against leptospirosis elicited by a live, attenuated lipopolysaccharide
dogs associated with leptospires. Am J Vet Res. 1979;40:839-844. mutant. J Infect Dis. 2011;203:870-879.
41. Geisen V, Stengel C, Brem S, et al. Canine leptospirosis infections— 63. Chandler AM, Goldstein RE. Assessing renal colonization in dogs
clinical signs and outcome with different suspected Leptospira 15 months after receiving a multi-serovar bacterin based on vac-
serogroups (42 cases). J Small Anim Pract. 2007;48:324-328. cine, vs unvaccinated dogs, all experimentally infected with Lep-
42. Sykes JE. Unpublished observations, 2012. tospira kirschneri serovar Grippotyphosa [Abstract]. J Vet Intern
43. Araujo ER, Seguro AC, Spichler A, et  al. Acute kidney injury in Med. 2010;24:764.
human leptospirosis: an immunohistochemical study with patho- 64. Klaasen HL, Molkenboer MJ, Vrijenhoek MP, et al. Duration of
physiological correlation. Virchows Arch. 2010;456:367-375. immunity in dogs vaccinated against leptospirosis with a bivalent inac-
44. Zaragoza C, Barrera R, Centeno F, et  al. Characterization of tivated vaccine. Vet Microbiol. 2003;95:121-132.
renal damage in canine leptospirosis by sodium dodecyl sulphate– 65. Monahan AM, Miller IS, Nally JE. Leptospirosis: risks during rec-
polyacrylamide gel electrophoresis (SDS-PAGE) and Western blot- reational activities. J Appl Microbiol. 2009;107:707-716.
ting of the urinary proteins. J Comp Pathol. 2003;129:169-178. 66. Stern EJ, Galloway R, Shadomy SV, et al. Outbreak of leptospiro-
45. Forrest LJ, O’Brien RT, Tremelling MS, et al. Sonographic renal findings sis among adventure race participants in Florida, 2005. Clin Infect
in 20 dogs with leptospirosis. Vet Radiol Ultrasound. 1998;39:337-340. Dis. 2010;50:843-849.
46. Barr SC, McDonough PL, Scipioni-Ball RL, et  al. Serologic 67. Strugnell BW, Featherstone C, Gent M, et al. Weil’s disease associ-
responses of dogs given a commercial vaccine against Leptospira ated with the adoption of a feral rat. Vet Rec. 2009;164:186.
interrogans serovar pomona and Leptospira kirschneri serovar 68. Martins G, Penna B, Lilenbaum W. The dog in the transmission of
grippotyphosa. Am J Vet Res. 2005;66:1780-1784. human leptospirosis under tropical conditions: victim or villain?
47. Miller MD, Annis KM, Lappin MR, et al. Variability in results of the Epidemiol Infect. 2012;140:207-208.
microscopic agglutination test in dogs with clinical leptospirosis and dogs
vaccinated against leptospirosis. J Vet Intern Med. 2011;25:426-432.
CHAPTER 51

Lyme Borreliosis
Jane E. Sykes

appear to be relatively resistant to development of disease.5


Overview of Lyme Borreliosis Dogs can develop fever, arthritis, and renal disease, but most
First Described: Connecticut, USA, 1982 (Burgdorfer)1 infected dogs show no signs of illness.
Cause: Borrelia burgdorferi sensu lato (a spirochete) More than 15 different species of B. burgdorferi sensu lato
have been described, some of which are nonpathogenic (Table
Primary Mode of Transmission: Ixodes spp. ticks
51-1). Borrelia turicatae, the cause of tick-borne relapsing fever
Affected Hosts: Humans and a large variety of animals; dis- in humans, has been detected in sick dogs from Texas.6 The
ease occurs in dogs, humans, horses, cattle DNA of Borrelia lonestari, which is thought to be nonpatho-
Geographic Distribution: North America, Europe, Asia genic, has been detected in dogs from Arkansas.7 In North
America, Lyme disease is caused by Borrelia burgdorferi sensu
Major Clinical Signs: Fever, lethargy, inappetence, lameness
stricto, whereas in Europe and Asia, other species that belong
due to polyarthritis. Signs of Lyme nephropathy include
to Borrelia burgdorferi sensu lato are more important, spe-
vomiting, weight loss, and polyuria and polydipsia.
cifically Borrelia afzelii and Borrelia garinii. This may account
Differential Diagnoses: Differential diagnoses for suspected for differences in clinical manifestations that occur in human
Lyme polyarthritis includes bilateral cruciate ligament rup- patients in Europe compared with those in North America (see
ture, primary immune-mediated polyarthritis, septic poly- Public Health Significance, later). Some strains of B. burgdorferi
arthritis, and polyarthritis secondary to infection with other appear to have increased virulence, such as B. burgdorferi OspC
pathogens such as Ehrlichia ewingii, Bartonella spp., Ehrlichia type A within the United States.8
canis, Anaplasma phagocytophilum, Rickettsia rickettsii, B. burgdorferi sensu lato is transmitted by Ixodes ricinus-
and fungal organisms. Differential diagnosis for suspected persulcatus complex ticks (Table 51-2). The geographic distri-
Lyme nephritis include leptospirosis, bacterial pyelone- bution of Lyme disease reflects that of the vector ticks as well
phritis, primary immune-mediated glomerulonephritis, as the competency of the reservoir hosts involved. In the United
familial nephropathies, amyloidosis, and glomerulonephri- States, the vectors are Ixodes scapularis in the east and upper
tis secondary to other chronic infections such as Dirofilaria Midwest, and Ixodes pacificus in the West (see Chapter 29).
immitis, Babesia canis, E. canis, and Leishmania spp. infections. Major foci of infection exist in the upper Midwest, Northeast,
Human Health Significance: Dogs and cats are not a direct mid-Atlantic, and parts of northern California (Figure 51-1).9
source of human infection but may bring unfed infected In some endemic areas, seroprevalence in dogs is nearly 90%,10
ticks into the house. Evidence of canine exposure to although seroprevalence in a study that used a C6 ELISA assay
B. burgdorferi is a sentinel for human exposure. (see Diagnosis, later) was lower than this in endemic areas such
as Connecticut and Massachusetts, around 20%.11 Infection is
passed transstadially within the tick (i.e., from larva to nymph
to adult), and not transovarially (from adult to egg). Reser-
Etiologic Agent and Epidemiology voirs for the spirochete in the Northeast and the upper Mid-
west are Peromyscus leucopus, the white-footed mouse, which
Lyme borreliosis, or Lyme disease, is a vector-borne spirochet- can harbor large numbers of the organism without overt signs
osis caused by the motile, corkscrew-shaped bacterium Borrelia of illness,12 as well as shrews and chipmunks.13 In the western
burgdorferi sensu lato. Lyme disease occurs in North America, United States, the western gray squirrel is the primary reser-
Europe, and Asia. It was named after Old Lyme in Connecticut, voir host.14 Lyme disease is less prevalent in the western United
where clusters of the disease were first recognized in children States because I. pacificus prefers to feed on the western fence
with juvenile rheumatoid arthritis in 1976.2 However, chronic lizard, a poor reservoir for the spirochete. Similarly, the low
cutaneous manifestations of infection were recognized in human prevalence of Lyme disease in the southeastern United States,
patients in Germany as far back as 1883.3 The spirochete was is because I. scapularis ticks feed primarily on lizards in this
first detected in ticks by Burgdorfer in 1982.1 A variety of differ- region.15 B. burgdorferi is primarily transmitted to humans
ent mammalian hosts and humans may be infected. The disease by nymphal ticks, because they are extremely small and often
has been increasingly recognized and is now the most common enough go unnoticed. Adult ticks may be more important for
vector-borne infectious disease of humans in the United States transmission of infection to dogs.16 The nymphs of I. scapularis
and Europe. Reforestation of farmland and proliferation of deer quest in the late spring and summer, when humans and dogs are
and tick populations may have contributed to emergence of the often outdoors and become exposed. The peak questing times
disease in humans.4 Cats can be infected and seroconvert, but for I. scapularis adult ticks are in the spring and fall. Other

487
488 SECTION 2  Bacterial Diseases

TABLE 51-1
Pathogenic and Nonpathogenic Species That Belong to Borrelia
burgdorferi sensu lato and Their Geographic Distributions
United States Europe Asia
Pathogenic B. burgdorferi B. burgdorferi B. afzelii
sensu stricto sensu stricto B. garinii
B. afzelii
B. garinii
Non-pathogenic B. bissettii B. valaisiana* B. bissettii*
or question- B. andersonii B. bissettii* B. japonica
able patho­ B. californiensis B. spielmanii* B. turdi FIGURE 51-1  Approximate geographic distribution of Lyme borreliosis in humans
genicity B. carolinensis B. lusitaniae B. tanukii (and dogs) in the United States.
B. americana B. bavariensis B. sinica
B. kurtenbachii B. yangtze

*B. valaisiana and B. bissettii have been isolated from single cases of
Lyme borreliosis; B. spielmanii has been detected in early skin lesions
(Stanek G, Reiter M. The expanding Lyme Borrelia complex—clinical
significance of genomic species? Clin Microbiol Infect 2011;17:487-493).

TABLE 51-2
Vectors of Lyme Disease Worldwide
Geographic Tick Common Reservoir host
Location Species Name for Borrelia
Western United Ixodes Pacific black- Western gray
States ­pacificus legged tick squirrel
Northeastern Ixodes Black-legged White-footed
and upper scapularis tick mouse, shrews,
­Midwestern ­chipmunks
United States
Europe Ixodes Castor bean Squirrels,
ricinus or sheep thrushes,
tick chipmunks,
mice, shrews,
hedgehogs,
rats, hares,
pheasants, FIGURE 51-2  Countries and regions where Lyme borreliosis occurs in Europe. The
voles size of the red dot correlates with the relative prevalence of the infection in those areas.
Asia Ixodes Taiga tick Voles
persulcatus region. A huge variety of reservoir hosts appear to be involved in
Europe. Small rodents such as squirrels appear to be an important
reservoir host for B. afzelii, and birds such as blackbirds and song
vector-borne pathogens, such as Anaplasma phagocytophilum, thrushes are important reservoir hosts for B. garinii.21
may be co-transmitted and complicate the clinical picture. There The complete genome of B. burgdorferi has been sequenced.
is some evidence that co-infection with A. phagocytophilum can The organism expresses different outer-surface lipoproteins at
enhance the pathogenicity of B. burgdorferi.17 different stages of infection, which allows it to adapt to dra-
In continental Europe, Lyme disease is most prevalent in cen- matically different environments within the arthropod vector
tral and eastern Europe, especially Poland, Slovakia, and Slovenia, and the mammalian host. This property of the organism has been
but also Germany, Switzerland, Austria, and southern Scandina- exploited for the purpose of diagnostic assay and vaccine devel-
via (Figure 51-2). B. garinii was detected in a dog from the Czech opment. Outer surface proteins of importance include OspA,
republic with meningoencephalitis,18 and B. afzelii has been OspC, and VlsE. Over the fall, winter, and spring, the spirochete
detected in dogs from Poland.19 Evidence of infection has also remains dormant within the nymphal tick and expresses OspA,
been found in dogs from the British Isles,20 where endemic areas which allows it to adhere to the tick midgut. When the tick ingests
include the Scottish Highlands, Ireland, Wales, the Lake District, mammalian blood in the late spring and summer, OspA expres-
the Yorkshire Moors, Exmoor, Wiltshire, Berkshire, and particu- sion is downregulated by the spirochete, and OspC expression is
larly the New Forest, the South Downs, and the Thetford Forest upregulated.22 The spirochete then moves to the tick hemolymph
CHAPTER 51  Lyme Borreliosis 489

and salivary glands. OspC binds a tick salivary gland protein,


which may help the organism evade the host immune response.
OspC also binds to mammalian plasminogen and helps the spiro-
chete to disseminate within the mammalian host.23 VlsE under-
goes recombinational shuffling of its genetic code, which further
allows the spirochete to evade the immune response.24 Finally,
B. burgdorferi may undergo metamorphosis into a spherical
shape when it encounters unfavorable conditions within the host,
such as when it is exposed to antibiotics, nutrient deprivation,
and changes in pH.25 This may also contribute to its ability to
evade the immune system and resist antimicrobial drug treatment.

Clinical Features
Signs and Their Pathogenesis
Transmission is thought to require tick attachment for a mini-
mum of 24 hours,26 but in some cases transmission may occur FIGURE 51-3  Borrelia burgdorferi in the metatarsal tendon sheath of a mouse with
at earlier time points.27 The spirochete replicates at the site severe combined immunodeficiency. (Courtesy Dr. Stephen Barthold, Center for Compara-
of tick attachment, then disseminates to multiple locations. tive Medicine, University of California, Davis.)
Although it can be transiently found in blood, the organism pri-
marily replicates and spreads through connective tissue (Figure with Lyme nephritis may have a thin body condition, dehydration,
51-3). It binds proteins such as plasminogen, β3 integrins such and show evidence of peripheral edema, pleural effusion, or asci-
as the platelet integrin αIIbβ3, glycosaminoglycans, fibronectin, tes, especially after crystalloid fluid administration. Complications
laminin, and decorin (a collagen proteoglycan).28 After inva- of hypertension can sometimes occur, such as retinal detachment.
sion, the organism can persist in dogs for over a year, through
evasion of host immune responses.29 Diagnosis
In contrast to human Lyme disease, dogs do not develop an early
skin rash (erythema migrans). It has been estimated that only 10% Diagnosis of Lyme disease is notoriously difficult. The over-
of affected dogs show overt signs of illness. Initial signs in dogs whelming majority of infected dogs experience subclinical infec-
occur 2 to 5 months after a tick bite and consist of variable fever, tion, after which positive antibody titers may persist for months
inappetence, thrombocytopenia, and lameness due to neutrophilic or years. Because only a small percentage (e.g., <10%) of infec-
polyarthritis. In a study of experimentally infected dogs, the first tions result in disease, it is important to distinguish between
joint to be affected was the joint closest to the site of the tick bite, diagnosis of infection or previous exposure to the spirochete and
which supports the notion that spirochetes reach the joints as a diagnosis of Lyme disease. Dogs with positive antibody titers may
result of spread through connective tissue.29,30 Subsequently, other develop unrelated illnesses, which results in diagnostic confusion.
joints can be affected, and dogs may develop shifting lameness. It As in human patients, the diagnosis must be established based on
is not clear whether dogs develop other clinical manifestations seen characteristic clinical signs, a history of exposure in an endemic
in humans such as persistent, antibiotic-refractory Lyme arthritis; area, and a positive antibody response to B. burgdorferi.39 A his-
uveitis; carditis; or encephalopathy. Complete heart block was tory of tick exposure may not always be present.
described in a seropositive dog from Connecticut that had patho-
logic changes at necropsy consistent with Lyme carditis.31 Laboratory Abnormalities
Lyme nephritis is a syndrome of membranoproliferative glo- Complete Blood Count
merulonephritis that has been recognized in dogs in association The most common finding on the CBC of dogs with Lyme poly-
with exposure to B. burgdorferi. Rare reports of a similar dis- arthritis or nephritis is mild to moderate thrombocytopenia.
ease in human patients exist.32 Golden and Labrador retriever The mechanism of thrombocytopenia is not known, but it may
breeds appear to be overrepresented,33 and it has been suggested be immune mediated, because treatment of dogs with Lyme
that Shetland sheepdogs and Bernese mountain dogs might also nephritis with immunosuppressive drugs can result in normal-
be predisposed.34-36 Dogs with Lyme nephritis are younger than ization of the platelet count. Mild to moderate anemia and leu-
dogs with other glomerular diseases. In one study, more than 50% kocytosis due to a mature neutrophilia may be present in dogs
of dogs with Lyme nephritis were 5 years of age or younger.33 with Lyme nephritis, but white cell counts may also be normal.
Many affected dogs are also thrombocytopenic, and some also Lymphopenia and mild neutropenia can also occur.
have polyarthritis.33 The pathogenesis of Lyme nephritis is uncer-
tain; immune-mediated mechanisms have been proposed.37 The Serum Biochemical Tests
DNA or antigen of the spirochete cannot be consistently detected Dogs with polyarthritis typically have minimal changes on serum
within renal biopsies.37,38 Subendothelial deposits of IgM, IgG, biochemistry testing. Dogs with Lyme nephritis may show azo-
and C3 can be detected within the glomeruli.33 Clinical signs temia, mild to marked hypoalbuminemia, metabolic acidosis,
result from proteinuria and renal failure and include inappetence, and electrolyte changes such as hyperphosphatemia, hypochlo-
lethargy, weight loss, vomiting, and polyuria and polydipsia. remia, and mild hyperkalemia or hypokalemia. Uncommonly,
hypercholesterolemia is present.
Physical Examination Findings
Physical examination findings in dogs with Lyme arthritis include Urinalysis
lethargy, fever, lameness, and swollen, warm, painful joints. Mild The urinalysis in dogs with Lyme nephritis generally reveals
generalized peripheral lymphadenopathy may be present. Dogs isosthenuria and proteinuria. Pyuria and microscopic hematuria
490 SECTION 2  Bacterial Diseases

may also be present. Urine protein to creatinine ratios are often Diagnostic Imaging
greater than 5 and may be above 15 in some dogs. Plain Radiography
Lyme arthritis is a nonerosive polyarthritis, so the only changes
Coagulation Profile visible on plain radiography of the joints are increased periar-
Dogs with Lyme nephritis may have low antithrombin activities ticular soft tissue opacity. Thoracic radiography is generally
as a result of glomerular antithrombin loss, which may lead to unremarkable, but pleural effusion is occasionally identified in
hypercoagulability syndromes. Sometimes, abnormalities such severely hypoalbuminemic dogs with Lyme nephritis.
as a shortened PT, prolonged APTT, and increased fibrinogen
and D-dimer concentrations are present.40 Sonographic Findings
Abdominal sonographic examination is normal in dogs with
Synovial Fluid Cytology Lyme arthritis. In dogs with nephritis, thickening and increased
Dogs with Lyme arthritis may have normal synovial fluid cytol- echogenicity of the renal cortices, decreased renal corticomedul-
ogy,30 or there may be markedly increased numbers of nonde- lary distinction, and peritoneal effusion may be seen.
generate neutrophils within the synovial fluid of distal joints
(>5000 and frequently >10,000 cells/µL). Because not all joints Microbiologic Tests
may be affected, synovial fluid should be collected from at least Specific diagnostic assays available for Lyme borreliosis in dogs
three and preferably four peripheral joints. are described in Table 51-3.

TABLE 51-3
Diagnostic Assays Available for Lyme Borreliosis in Dogs
Assay Specimen Type Target Performance
Bacterial isolation Skin biopsy close to B. burgdorferi spirochete Low sensitivity, requires special media, and may take sev-
the tick bite site; eral weeks. Isolation does not imply that B. burgdorferi
synovial fluid is the cause of disease.
Serology (C6 assay) Serum Antibodies against Positive serology does not equate with Lyme disease,
B. burgdorferi C6 so test results must be interpreted in light of clinical
protein findings. False positives can occur in regions of low
prevalence. False negatives are rare, because antibodies
are present by the time dogs develop illness. Cross-­
reactivity with vaccine antibodies does not occur.
Serology (whole cell Serum Antibodies against As for C6 serology except that cross-reactivity with vac-
IFA and ELISA) B. burgdorferi antigens cine antibodies occurs, and false positives may also
occur with other inflammatory diseases, and other
spirochete infections.
Serology (Western Serum Antibodies against Used to confirm the serological response to natural infec-
immunoblot) B. burgdorferi antigens tion in dogs that test positive with whole-cell ELISA
and IFA. Technically difficult to perform and requires
expertise to interpret. Positive serology does not equate
with Lyme disease, so test results must be interpreted in
light of clinical findings.
Serology (multiplex Serum Antibodies against Antibodies to OspC appear only early in infection, where-
fluorescent bead B. burgdorferi OspA, as those to OspF reflect chronic infection and appear
assay) OspC, and OspF to correlate with the C6 antibody response. Sensitivity
and specificity in dogs with naturally occurring Lyme
disease unknown. Positive serology does not equate
with Lyme disease, so test results must be interpreted in
light of clinical findings.
Serology (multi- Serum Antibodies against Sensitivity and specificity in dogs with naturally occurring
target silicon disc– B. burgdorferi OspA, Lyme disease requires further study. Positive serology
based assay) OspC, and OspF, SLP, does not equate with Lyme disease, so test results must
and P39 be interpreted in light of clinical findings.
PCR As for isolation B. burgdorferi DNA Rapid but insensitive. Synovial fluid from dogs with
polyarthritis may be the best specimen, but further
study is needed. Assay performance can vary between
laboratories.
CHAPTER 51  Lyme Borreliosis 491

Culture A multiplex fluorescent bead assay has been marketed in


B. burgdorferi can be isolated from tissue specimens in Barbour- North America for detection of antibodies to three antigens
Stoenner-Kelly medium. The optimal specimen for culture is a of B. burgdorferi: OspA, OspC, and OspF.43 The assay uses
skin biopsy collected from a site adjacent to the tick bite, which tiny beads to which OspA, OspC, and OspF are coupled. Dog
is rarely identifiable in affected dogs. Incubation of cultures for serum is added to the beads, and if present, antibodies in the
several weeks may be required. Because of these factors, culture serum bind to the antigens and can be detected using a fluores-
is not generally used on a routine basis for diagnosis. cent conjugate. The pattern of reactivity to each antigen can be
used to differentiate among the response to vaccination, early
Serologic Diagnosis infection, and chronic infection. The presence of anti-OspA
Serologic tests for antibody are the main assays used for diagno- antibodies suggests previous vaccination, because vaccines
sis of Lyme disease in humans and also in dogs, but it is critical contain OspA, and the spirochete rarely expresses OspA within
to recognize that positive serology does not necessarily equate
with the presence of Lyme disease. In Europe, infection with

Vaccination  infection
nonpathogenic variants of B. burgdorferi sensu lato can result in

Vaccination rOspA
positive serologic results, which further complicates diagnosis.

Vaccination lysate
Because the infection is chronic and persistent, and the incuba-

Control OspA
tion period is long, paired serology is generally not performed,
because seroconversion may not occur. However, paired serol-

Negative

Infection
Control
ogy using serologic panels that include B. burgdorferi may be
required for diagnosis of other vector-borne diseases that might
be present. Positive test results in regions of low prevalence are
more likely to be false positives than in regions of high preva-
lence, so interpretation of positive results should be performed
with care in low-prevalence regions.
Markline
Currently, one of the most widely used serodiagnostic tests for Serum control
canine Lyme disease is based on a C6 ELISA, which detects anti- lgG
bodies against a portion of the VlsE lipoprotein. The advantages Conjugate
lgA
control
of the C6 ELISA assay are that (1) it detects IgG antibodies 3 to lgM
5 weeks after the time of infection, so by the time dogs develop VlsE
clinical signs they are virtually always seropositive,41 and (2) it
is negative in dogs that have been vaccinated for Lyme disease,
because the antigen is not expressed by organisms used in Lyme P83/100
vaccines. The C6 ELISA is available as an in-practice lateral-flow
assay, in combination with serodiagnostic spots for Ehrlichia
canis/Ehrlichia ewingii antibody, Anaplasma spp. antibody, and
p58
Dirofilaria immitis antigen (SNAP 4Dx Plus, IDEXX Labora-
tories, ME) and as a quantitative ELISA (Quant C6), which is
performed at IDEXX central veterinary diagnostic laboratories.
p43
There is no correlation between the magnitude of the C6 ELISA
titer and disease severity. In a study from Europe, positive test FlaB p41
results did not correlate with disease.36 Another rapid immuno- BmpA p39
chromatographic ELISA assay has recently become available in
the United States (Abaxis VetScan Canine Lyme Rapid Test) that
OspA
detects antibodies to a different portion of the VlsE lipoprotein,
OspC and p41; these are combined on a single line. p30
Other serologic assays that are available include whole-
cell ELISA or immunofluorescent antibody (IFA) assays and OspC
Western blotting (WB; see Chapter 2 for a description of these
techniques). WB has traditionally been considered the gold
standard assay. False positives using whole-cell ELISA and p21
IFA have the potential to occur in patients with other spiro-
chete infections, with other inflammatory disorders, and in Osp17
vaccinated dogs. In human patients, a two-tiered approach is
recommended for diagnosis.39 Serum from patients who test
positive for IgM or IgG with ELISA is then subjected to WB,
because WB has increased specificity. WB has been used to dif- FIGURE 51-4  Western immunoblots of sera from dogs not infected with B. burgdor-
feri (Negative); immunized with recombinant OspA (vaccination rOspA); Immunized with a
ferentiate the response to immunization and natural infection
whole cell lysate vaccine (vaccination lysate); immunized and infected with B. burgdorferi
in dogs (Figure 51-4).42 It has also been used to identify “dual (vaccination + infection); and infected only with B. burgdorferi (infection). The left strip
status” dogs, that is, dogs that have been both immunized and shows all major signals available on the blot (control); the second strip from the left is
naturally infected. WB is more time-consuming to perform stained with a monoclonal antibody against OspA (control OspA). (Courtesy R. Straubinger,
than ELISA and IFA and experience is required to interpret it Ludwig-Maximillians-Universitat, Munich, Germany. In: Greene CE: Infection Diseases of
correctly.43 the Dog and Cat. 4th ed. St. Louis: Elsevier/Saunders; 2012.)
492 SECTION 2  Bacterial Diseases

the host. OspC is expressed as the spirochete moves to the tick


salivary glands and shortly after it enters the host. Thus an
antibody response to OspC may suggest recent infection; titers

Antibody titer
decline and become undetectable beyond 3 months after infec- C6
tion.44 Antibodies to OspC appear as early as 3 weeks after OspF
experimental infection of dogs, and OspF as early as 5 weeks.44
OspF is expressed in more chronic infections, and can be
detected together with the C6 antibody response in naturally
exposed dogs (Figure 51-5).44,45 In dogs, when WB was used OspC
as the gold standard, the sensitivities of the OspA, OspC, and Infection 3mon
OspF assays were 83%, 62%, and 82%, and the specificities Time
were 90%, 89%, and 86%, respectively. The use of WB as FIGURE 51-5  Proposed canine antibody response to OspC, OspF, and C6 antigens
the gold standard was questioned, and it was suggested that of B. burgdorferi during early and late infection. Data were obtained by fluorescent bead
in fact, the fluorescent bead assay may have greater sensitivity multiplex analysis. The lines for the first 3 months after infection are based on multiplex
results from experimentally infected dogs. After 3 months the lines are projected from
and specificity than WB. The performance of the fluorescent
data obtained from patient sera. The horizontal dotted line shows the cutoff value for the
bead assay is yet to be thoroughly evaluated in dogs with natu- multiplex assay. The vertical dotted line indicates 3 months after infection. (Modified from
rally occurring Lyme disease. Wagner B, Freer H, Rollins A, et al. Antibodies to Borrelia burgdorferi OspA, OspC, OspF,
A novel silicon disc–based serologic assay is available in and C6 antigens as markers for early and late infection in dogs. Clin Vaccine Immunol
the United States for detection of antibodies to B. burgdorferi, 2012;19:527-535.)
E. canis, and A. phagocytophilum and antigen to Dirofilaria
immitis (Accuplex 4, Antech Diagnostics, Irvine, CA). The assay microscopy.48 Other findings in experimentally infected dogs
for B. burgdorferi detects antibodies to the spirochete proteins include periarteritis and perineuritis, especially within joint
OspA, OspC, OspF, P39, and SLP and is performed at central capsules and the skin, as well as lymphoid hyperplasia within
veterinary diagnostic laboratories. Preliminary data suggests peripheral lymph nodes. Mild, focal meningitis and encephalitis
that laboratory analysis of the response to these proteins cor- was identified in dogs that were infected with B. burgdorferi
relates with the results of WB and can differentiate between the and immunosuppressed with dexamethasone.49
responses to natural infection and vaccination, and acute and Renal histopathology in dogs with Lyme nephritis reveals dif-
chronic infection.46 fuse membranoproliferative glomerulonephritis (Figure 51-6),
dilation of the cortical renal tubules, tubular necrosis and regen-
Molecular Diagnosis Using the Polymerase Chain Reaction eration, and mild to moderate, diffuse interstitial lymphoplas-
As with culture, PCR assays are best performed on skin biopsy macytic inflammation.33 Periglomerular or diffuse interstitial
specimens collected from a region adjacent to the tick bite site. fibrosis may be present in dogs with end-stage disease.
In human patients, the use of PCR assays for diagnosis of Lyme
borreliosis on blood has low sensitivity, and so false negatives are Treatment and Prognosis
common when B. burgdorferi PCR assay is used as part of a whole-
blood vector-borne infection PCR panel. Synovial fluid or synovial Antimicrobial Treatment
membrane biopsies may be the optimum specimen for diagnosis Antibiotic treatment is recommended for seropositive dogs that
of Lyme arthritis in dogs. This is also true in human patients.47 have clinical illness consistent with Lyme disease. There is no
However, in a study of experimentally infected dogs, PCR assay of evidence that treatment of healthy seropositive dogs is beneficial
the synovial fluid was insensitive.30 The sensitivity and specificity and it may lead to drug adverse effects and contribute to antimi-
of PCR assays when used on specimens such as synovial fluid for crobial resistance in other bacteria and antimicrobial shortages.
diagnosis of canine Lyme borreliosis requires further study. The antibiotic of choice for Lyme arthritis is doxycycline. The
optimal dose and duration of treatment is unknown. Recom-
Pathologic Findings mended doses have included 5 mg/kg PO q12h and 10 mg/kg
Gross Pathologic Findings PO q12h or 10 mg/kg PO q24h.34,50 Four weeks of treatment
Gross pathologic findings in dogs with Lyme arthritis include has been recommended34 because the clinical manifestations of
peripheral lymphadenomegaly, joint swelling, and synovial disease resemble those of late-stage disease in human patients,
effusion.29 The synovial fluid may be yellow-tinged and cloudy, for which relapses occur when treatment durations of less than
with decreased viscosity. In dogs with nephritis, the kidneys 30 days are used.39 Dogs with polyarthritis generally respond
are diffusely light tan and may have pinpoint red foci over the clinically to doxycycline treatment within 24 to 48 hours. Other
cortical surfaces.33 In addition, evidence of systemic edema, differential diagnoses for polyarthritis should be considered if
thrombosis and infarction, and uremia (such as parathyroid an inadequate response to treatment occurs. On the other hand,
hyperplasia, ulcerative stomatitis, and gastritis) may be present. a clinical response to doxycycline treatment is not sufficient to
make a diagnosis of Lyme arthritis. This is because there are
Histopathologic Findings other doxycycline-responsive causes of infectious polyarthritis
Histopathology of the joints of dogs that have been experimen- in dogs; signs of primary immune-mediated polyarthritis can
tally infected with B. burgdorferi reveals fibrinosuppurative or wax and wane; and doxycycline has antiinflammatory proper-
lymphoplasmacytic inflammation of the synovial membranes, ties that may contribute to clinical improvement. For dogs that
joint capsules, and tendon sheaths.29,30 Inflammation can some- do not tolerate doxycycline, amoxicillin can be used, which also
times be found in the joints even when a history of lameness has activity against B. burgdorferi (Table 51-4). Azithromycin
is not present.29 In human patients, organisms have been dem- and third-generation cephalosporins have also been used to
onstrated in the synovium using immunostains and electron treat Lyme disease in human patients.39 Doxycycline treatment
CHAPTER 51  Lyme Borreliosis 493

TABLE 51-4
Suitable Antimicrobials for Treatment of Lyme Disease in Dogs
Interval Duration
Drug Dose Route (hours) (days)
Doxycycline 5 to 10 mg/kg PO 12 28
Amoxicillin 20 mg/kg PO 8 28

TABLE 51-5
Immunosuppressive Drug Protocols That Could Be Considered
for Treatment of Lyme Nephritis in Dogs in Conjunction with
Antimicrobial Drug Treatment
FIGURE 51-6  Membranoproliferative glomerulonephritis in a 6-year-old, intact
male golden retriever dog that was seropositive for B. burgdorferi C6 protein antibodies Interval and
and had clinical and biochemical evidence of protein-losing nephropathy. The morpho- Drug Dose Route Duration
logic diagnosis was moderate to severe, diffuse, global membranoproliferative glomeru- Methylprednisolone 5 mg/kg IV 24 hours for
lonephritis with moderate chronic-active tubulointerstitial nephritis. The capillary walls sodium ­succinate 2 days
are thickened and there is mesangial cell proliferation. There was also evidence of focal with either
arteriolar mural disorganization, lamination, and sclerosis in this biopsy specimen. (Image
cyclophosphamide,
courtesy Dr. George Lees, Texas A&M University.)
mycophenolate
mofetil, or aza-
is also recommended for dogs with Lyme nephritis, although thioprine below
clinical improvement generally does not occur with antimicro- Cyclophosphamide 200 mg/m2 IV Every 14 days for
bial treatment alone (see Supportive Care). a maximum
Although treatment leads to clinical improvement and a reduc- of 6 cycles.
tion in antibody titers, B. burgdorferi can persist in tissues after Recheck CBC
treatment is discontinued. Studies in mouse and primate models 1 week after
have shown that despite negative cultures after treatment, PCR each treatment.
assays remain positive, and infection can be transmitted from
Mycophenolate 10 mg/kg IV or 12 hours until
PCR-positive to naïve animals.51,52 Regardless of the serologic
mofetil PO ­remission occurs,
test used, antibody titers can persist for months or years after res-
then consider
olution of clinical signs and antimicrobial drug treatment, so the
tapering
results of antibody testing are not consistently useful to guide fur-
ther treatment when clinical resolution of arthritis has occurred. Azathioprine 1-2 mg/kg PO 24 hours for 7
days, then every
Supportive Care 48 hours until
Dogs with arthritis usually respond rapidly to treatment with anti- remission occurs,
biotics, and additional treatment may not be necessary. Severe pain then consider
associated with arthritis may be treated with nonsteroidal antiin- tapering
flammatory drugs or opiate analgesics such as tramadol. Because
Lyme arthritis can be reactivated in some dogs by administration
of glucocorticoids more than a year after exposure,53 glucocor-
ticoids are not recommended for their antiinflammatory effects. Prognosis
Dogs with Lyme nephritis require management for protein- It is estimated that more than 90% of dogs that are infected
losing nephropathy, which may include treatment with intrave- with B. burgdorferi show no signs of illness. Dogs with Lyme
nous crystalloids and colloids, angiotensin-converting enzyme arthritis generally recover rapidly with antimicrobial treatment
inhibitors such as enalapril or benazepril, low-dose aspirin in an and do not develop relapse of disease. It is not clear whether
attempt to reduce thrombotic events, and nutritional support the “antibiotic-refractory arthritis” that occurs in a small per-
with a reduced protein diet. Placement of an esophagostomy or centage of genetically predisposed human patients also occurs in
gastrostomy tube for feeding purposes may be required. Blood dogs. Seropositive dogs with antibiotic-refractory polyarthritis
pressure should be monitored and if necessary, antihypertensive may have other unrelated causes of their disease, such as pri-
drugs such as amlodipine should be administered and titrated to mary immune-mediated polyarthritis.
effect. Anecdotally, improved outcomes have been noted in dogs The prognosis for dogs with Lyme nephritis is guarded to
with Lyme nephritis after immunosuppressive drug treatment poor. In the past, it was noted that most dogs die or are eutha-
(Table 51-5). The optimum protocol is yet to be determined. nized within days to weeks.34 Death often results from systemic
Dogs treated with immunosuppressive drugs should be carefully thrombosis or oliguric or anuric renal failure. Anecdotally, lon-
monitored for adverse effects of drug therapy. ger survival times of months to over a year have been noted in
494 SECTION 2  Bacterial Diseases

some dogs treated with immunosuppressive drugs in addition to exposed individuals may offer some benefit, because the protec-
doxycycline and other supportive treatments. tion induced by vaccination (anti-OspA antibodies) differs from
that induced by natural infection (no anti-OspA antibodies).
Immunity and Vaccination However, whether ­recurrent Lyme disease occurs in dogs as a
result of reinfection is not known.
Borrelia spp. can evade the host immune response through mod-
ification of outer surface proteins and possibly metamorphosis Prevention
to a resistant spherical form.54 Interference with B-cell responses
by the spirochete also seems to occur.55 Humoral immunity Avoidance of tick-infested areas, use of topical ectoparasiti-
is most critical for resolution of infection, although T-cell cides, and routine inspection of dogs for ticks after outdoor
responses may be important for resolution of cell-mediated con- activities can help to prevent Lyme disease. Ticks should be
sequences of infection, such as carditis in human patients.56 removed within 24 hours of attachment, before transmission
Several vaccines are available in North America for reduction of the spirochete can occur, but removal up to 60 hours after
of Lyme disease in dogs. Lyme vaccines stimulate the formation of attachment may still reduce the chance of transmission.65 Refer
borreliacidal antibodies that are directed against the surface pro- to Chapter 28 for general information on tick prevention and
teins normally expressed by the spirochete when it resides within removal. Although treatment with a single dose of doxycycline
the tick. When the tick ingests dog blood that contains these anti- after a known Ixodes tick bite can prevent Lyme disease in
bodies, complement-mediated lysis of the bacteria occurs within humans66 and mice,67 it is controversial because of the very low
the tick. Thus, bacteria are inactivated before they invade the host. risk of infection after a tick bite.68 A study in mice showed that
A canine recombinant OspA vaccine is available that induces the treatment 2 or more days after tick removal was ineffective.67
formation of antibodies only against OspA. This subunit vaccine For healthy dogs that test positive for antibodies to B. burg-
is similar to a Lyme disease vaccine that was previously available dorferi during a heartworm screen with assays that include a
for prevention of human Lyme borreliosis (LYMErix, Glaxo- B. burgdorferi ELISA, a urinalysis could be offered in order
SmithKline). In humans, the vaccine was shown to be safe and to assess for proteinuria.34 If proteinuria is detected, further
efficacious, conferring immunity on 76% of adults and 100% work-up that includes a urine protein to creatinine ratio, aero-
of children with a low prevalence of local injection site reactions bic bacterial culture of the urine, serum biochemistry panel,
and flu-like symptoms.57 However, this vaccine was withdrawn and imaging may be indicated. However, whether treatment
in February 2002 because of concerns that it may trigger autoim- of seropositive, proteinuric dogs with doxycycline influences
mune disease as a result of molecular mimicry in sensitive individ- the progression of Lyme nephropathy is not known, so this
uals, despite a lack of scientific justification for these concerns.58,59 is controversial. Similarly, a CBC or platelet count could be
Two inactivated whole spirochete vaccines are also avail- performed, but whether treatment of thrombocytopenic dogs
able for dogs. One contains two strains of B. burgdorferi, one is necessary is also unknown. At a minimum, tick control for
of which expresses high levels of OspC. The other vaccine is seropositive, apparently healthy dogs should be recommended.
a single-strain vaccine. The potential advantage of these vac-
cines is that they stimulate immunity not only to OspA, but Public Health Aspects
also to other surface proteins expressed by the spirochete. This
may provide the opportunity to neutralize B. burgdorferi when Lyme disease is the most common vector-borne disease of
OspA is downregulated, such as when the organism is in the humans in the Northern Hemisphere, and the prevalence of the
salivary glands and after it enters the host. Concern has been disease has increased progressively.69 In humans, the first sign
expressed that whole-cell vaccines may be more likely to trig- of disease in 80% to 90% of infected individuals is erythema
ger autoimmune consequences in dogs than the subunit vaccine, migrans, which is a characteristic “bull’s-eye” rash that occurs
especially in dogs that have been previously exposed to B. burg- 3 to 30 days after a tick bite and moves outward from the bite
dorferi.34 However, no strong evidence exists that these types site at a rate of approximately 1 cm/day. In some people, the
of adverse reactions occur. Both OspA and whole-cell vaccines rash is pruritic or painful, and it may be accompanied by head-
provide protection against infection and disease that results ache and malaise. Early disseminated disease can appear as mul-
from experimental challenge.60,61 A study in an endemic area tiple erythema migrans rashes; myocardial disease that is usually
for Lyme disease found a reduced prevalence of C6 seropositiv- characterized by atrioventricular block; or neuroborreliosis.
ity in dogs that had been vaccinated with a whole-cell bacterin Neuroborreliosis is more common in Europe and may be char-
when compared with dogs that had not been vaccinated, sug- acterized by the development of cranial nerve palsies or signs of
gesting the possibility of protection from natural infection.62 meningitis and polyradiculoneuritis. Arthritis is a manifestation
The dual-strain bacterin has been shown to have a 1-year dura- of late Lyme disease. A small percentage of infected individuals,
tion of immunity.63 Although no Lyme vaccine can be relied especially those of certain human leukocyte antigen (HLA) types,
on to provide complete protection, side-by-side comparisons of can develop chronic, antibiotic-refractory arthritis, which may
canine Lyme vaccines that are available on the market have not result from persistence of spirochete residues in tissues.70 Euro-
been performed. In addition, whether Lyme vaccination protects pean strains, particularly B. afzelii, can also induce a chronic skin
against, or contributes to, the most severe consequence of infec- manifestation known as acrodermatitis chronica atrophicans.71
tion, Lyme nephritis, is unknown. Finally, whether there are Dogs do not pose a direct zoonotic risk to humans in the
any advantages or disadvantages of vaccinating dogs that are household. However, they may carry infected, unfed ticks
already seropositive as a result of natural infection remains to be into the household that could subsequently attach to humans.
elucidated. The recent identification of reinfection (as opposed The presence of antibodies in dogs can also indicate increased
to relapse) as a cause of recurrent clinical signs of borreliosis human risk as a result of common exposure to infected ticks in
in human patients64 suggests that vaccination of previously the environment (sentinel exposure).
CHAPTER 51  Lyme Borreliosis 495

CASE EXAMPLE Neutrophils 15,561 cells/µL (3000-10,500 cells/µL)


Lymphocytes 171 cells/µL (1000-4000 cells/µL)
Monocytes 1026 cells/µL (150-1200 cells/µL)
Signalment: “Dako”, a 7-year-old MC Doberman Pinscher dog Eosinophils 342 cells/µL (0-1500 cells/µL), platelets clumped.
from Amador County in northern California Serum Chemistry Profile:
History: Dako was evaluated for a 2-day history of lethargy, Sodium 148 mmol/L (143-151 mmol/L)
apparent blindness, inappetence, and inability to walk. He Potassium 3.5 mmol/L (3.6-4.8 mmol/L)
was taken to a local emergency clinic within 24 hours of Chloride 115 mmol/L (108-116 mmol/L)
illness, where a neurologic examination was considered Bicarbonate 20 mmol/L (20-29 mmol/L)
abnormal and an intracranial lesion was suspected. He was Phosphorus 3.1 mg/dL (2.6-5.2 mg/dL)
treated with intravenous fluids and given a single dose of Calcium 10.0 mg/dL (9.6-11.2 mg/dL)
ampicillin (1 g). The following day his mentation and thoracic BUN 11 mg/dL (11-33 mg/dL)
limb strength had improved, but he remained unable to Creatinine 0.8 mg/dL (0.8-1.5 mg/dL)
walk and developed urinary incontinence. He lived on a Glucose 102 mg/dL (86-118 mg/dL)
farm and had contact with sheep, horses, llamas, chickens, Total protein 6.1 g/dL (5.4-6.9 g/dL)
geese, cats, and four other dogs, all of which were currently Albumin 3.6 g/dL (3.4-4.3 g/dL)
healthy, although an additional dog had died of acute Globulin 2.5 g/dL (1.7-3.1 g/dL)
renal failure 1 month earlier. There was no travel history or ALT 17 U/L (21-72 U/L)
toxin exposure, but frequent exposure to ticks occurred. AST 36 U/L (20-49 U/L)
Dako’s diet consisted of commercial dry dog food, and ALP 60 U/L (14-91 U/L)
occasionally pieces of cooked steak and raw goose eggs. He Creatine kinase 217 U/L (55-257 U/L)
had not received a Lyme vaccine in the past but had been GGT 1 U/L (0-6 U/L)
vaccinated regularly for distemper, adenovirus, parvovirus, Cholesterol 167 mg/dL (139-353 mg/dL)
parainfluenza, and rabies. Dako had been diagnosed several Total bilirubin 0.1 mg/dL (0-0.2 mg/dL)
years previously with color dilution alopecia. Magnesium 1.8 mg/dL (1.9-2.5 mg/dL).
Current Medications: Monthly topical flea and tick Urinalysis: SGr 1.018; pH 8.0, negative protein (SSA), negative
preventative (fipronil and S-methoprene), monthly oral bilirubin, negative glucose, 0-1 WBC/HPF, 0-2 RBC/HPF.
heartworm preventative (ivermectin and pyrantel). Imaging Findings:
Physical Examination: Thoracic Radiographs: Unremarkable.
Body Weight: 43.1 kg Abdominal Ultrasound: The liver was diffusely hypoechoic but
General: Quiet, alert, and responsive. Hydrated. Ambulatory was normal in size. There was mild bilateral adrenomegaly
on all four limbs but appeared very painful. T = 101.9°F (0.9 cm). The urinary bladder was very large and was in a
(38.8°C), HR = 78 beats/min, RR = 24 breaths/min, mucous pelvic location.
membranes pink, CRT = 1 s. Microbiologic Testing: 4Dx SNAP test (IDEXX Laboratories):
Integument, Eyes, Ears, Nose, and Throat: A thin haircoat Positive for B. burgdorferi antibodies; negative for antibodies
with scaling was noted. Mild episcleral injection was present to Anaplasma spp. and E. canis; negative for Dirofilaria
bilaterally. Moderate dental calculus and gingivitis were also immitis antigen.
present.
Musculoskeletal: Body condition score was 5/9 and the dog Cytology of Synovial Fluid Obtained via Arthro­centesis
was symmetrically well muscled. Multiple distal joints were
severely swollen and painful on palpation, including both Right Left Right Left
carpi, tarsi, stifle, and elbow joints. The dog appeared painful Carpus Carpus Tarsus Stifle
when rising from recumbency and when walked, with
pronounced right pelvic limb lameness. Cell count 23,730 29,370 ND 1020
Cardiovascular, Respiratory, Gastrointestinal, and (cells/µL)
Genitourinary: No clinically significant abnormalities were Neutrophils 85 94 76 12
detected. The dog had a large bladder, and actively urinated (%)
a large amount during the examination. Rectal examination Small mono- 2 0 1 41
was unremarkable. nuclear
Lymph Nodes: Bilateral popliteal lymphadenomegaly (3 cm in cells (%)
diameter) was noted. The remaining peripheral lymph nodes
measured 2 cm in diameter. All nodes were soft on palpation. Large mono- 13 6 23 47
Neurologic Examination: No neurologic abnormalities were nuclear
detected. cells (%)
Laboratory Findings: Interpreta- Marked purulent inflam- Very mild puru-
CBC: tion mation with nondegen- lent inflam-
HCT 48.4% (40%-55%) erate neutrophils mation with
MCV 69.6 fL (65-75 fL) nondegenerate
MCHC 34.9 g/dL (33-36 g/dL) neutrophils
WBC 17,100 cells/µL (6000-13,000 cells/µL)
ND, not done.
496 SECTION 2  Bacterial Diseases

Lyme C6 Quant antibody ELISA (IDEXX Laboratories): 153 U/mL the course of illness for this dog to have seroconverted to
(normal, <30 U/mL). other vector-borne pathogens such as A. phagocytophilum.
PCR for B. burgdorferi on synovial fluid: Negative. Other diagnostic assays that might have been useful in this
Diagnosis: Neutrophilic polyarthritis; possibly secondary to dog (and in this geographic location) include serology for
B. burgdorferi infection. Bartonella spp.; PCR on blood for vector-borne pathogens
Treatment: Dako was treated with doxycycline (5 mg/kg PO such as A. phagocytophilum, Rickettsia spp., and Ehrlichia
q12h) and tramadol (2 mg/kg PO q8h) and showed dramatic canis; aerobic bacterial culture of the synovial fluid; and
clinical improvement within 24 hours of initiating treatment. convalescent serology for vector-borne pathogens. The
A physical examination was normal 2 weeks later. negative synovial fluid PCR result did not rule out the
Comments: The positive serology and response to possibility of Lyme arthritis, because organism numbers
doxycycline suggested the possibility of Lyme arthritis, within the synovial fluid of animals with Lyme arthritis may
although it is possible there was some other cause that was be extremely low. The dog had been treated with ampicillin
not identified and the presence of C6 antibodies was not before it was seen, which also may have reduced organism
related to this dog’s illness. It may have been too early in numbers further.

SUGGESTED READINGS
13. Brisson D, Dykhuizen DE, Ostfeld RS. Conspicuous impacts of
Little SE, Heise SR, Blagburn BL, et  al. Lyme borreliosis in dogs and inconspicuous hosts on the Lyme disease epidemic. Proc Biol Sci.
humans in the USA. Trends Parasitol. 2010;26(4):213-218. 2008;275:227-235.
Littman MP, Goldstein RE, Labato MA, et  al. ACVIM small animal 14. Salkeld DJ, Lane RS. Community ecology and disease risk: lizards,
consensus statement on Lyme disease in dogs: diagnosis, treatment, squirrels, and the Lyme disease spirochete in California, USA. Ecol-
and prevention. J Vet Intern Med. 2006;20(2):422-434. ogy. 2010;91:293-298.
Radolf JD, Caimano MJ, Stevenson B, et  al. Of ticks, mice and men: 15. Oliver Jr JH, Lin T, Gao L, et al. An enzootic transmission cycle of
understanding the dual-host lifestyle of Lyme disease spirochaetes. Lyme borreliosis spirochetes in the southeastern United States. Proc
Nat Rev Microbiol. 2012;10(2):87-99. Natl Acad Sci U S A. 2003;100:11642-11645.
16. Little SE, Heise SR, Blagburn BL, et  al. Lyme borreliosis in dogs
REFERENCES and humans in the USA. Trends Parasitol. 2010;26:213-218.
1. Burgdorfer W, Barbour AG, Hayes SF, et al. Lyme disease—a tick- 17. Nyarko E, Grab DJ, Dumler JS. Anaplasma phagocytophilum–infected
borne spirochetosis? Science. 1982;216:1317-1319. neutrophils enhance transmigration of Borrelia burgdorferi across the
2. Steere AC, Malawista SE, Snydman DR, et al. Lyme arthritis: an human blood brain barrier in vitro. Int J Parasitol. 2006;36:601-605.
epidemic of oligoarticular arthritis in children and adults in three 18. Kybicova K, Schanilec P, Hulinska D, et al. Detection of Anaplasma
Connecticut communities. Arthritis Rheum. 1977;20:7-17. phagocytophilum and Borrelia burgdorferi sensu lato in dogs in the
3. Buchwald A. Ein Fall von diffuser idiopathischer haut Atrophie. Czech Republic. Vector Borne Zoonotic Dis. 2009;9:655-661.
Arch Derm Syph. 1883;15:553-556. 19. Zygner W, Gorski P, Wedrychowicz H. Detection of the DNA of
4. Barbour AG. Fall and rise of Lyme disease and other Ixodes tick- Borrelia afzelii, Anaplasma phagocytophilum and Babesia canis in
borne infections in North America and Europe. Br Med Bull. blood samples from dogs in Warsaw. Vet Rec. 2009;164:465-467.
1998;54:647-658. 20. Shaw SE, Binns SH, Birtles RJ, et al. Molecular evidence of tick-
5. Burgess EC. Experimentally induced infection of cats with Borrelia transmitted infections in dogs and cats in the United Kingdom. Vet
burgdorferi. Am J Vet Res. 1992;53:1507-1511. Rec. 2005;157:645-648.
6. Whitney MS, Schwan TG, Sultemeier KB, et  al. Spirochetemia 21. Hanincová K, Taragelová V, Koci J, et al. Association of Borrelia
caused by Borrelia turicatae infection in 3 dogs in Texas. Vet Clin garinii and B. valaisiana with songbirds in Slovakia. Appl Environ
Pathol. 2007;36:212-216. Microbiol. 2003;69:2825-2830.
7. Fryxell RT, Steelman CD, Szalanski AL, et al. Survey of Borreliae 22. Pal U, Yang X, Chen M, et al. OspC facilitates Borrelia burgdor-
in ticks, canines, and white-tailed deer from Arkansas. U.S.A. Para- feri invasion of Ixodes scapularis salivary glands. J Clin Invest.
sit Vectors. 2012;5:139. 2004;113:220-230.
8. Strle K, Jones KL, Drouin EE, et  al. Borrelia burgdorferi RST1 23. Lagal V, Portnoi D, Faure G, et  al. Borrelia burgdorferi sensu
(OspC type A) genotype is associated with greater inflammation stricto invasiveness is correlated with OspC-plasminogen affinity.
and more severe Lyme disease. Am J Pathol. 2011;178:2726-2739. Microbes Infect. 2006;8:645-652.
9. Carrade D, Foley J, Sullivan M, et al. Spatial distribution of sero- 24. Dresser AR, Hardy PO, Chaconas G. Investigation of the genes
prevalence for Anaplasma phagocytophilum, Borrelia burgdorferi, involved in antigenic switching at the vlsE locus in Borrelia burg-
Ehrlichia canis, and Dirofilaria immitis in dogs in Washington, dorferi: an essential role for the RuvAB branch migrase. PLoS Pat-
Oregon, and California. Vet Clin Pathol. 2011;40:293-302. hog. 2009;5:e1000680.
10. Magnarelli LA, Anderson JF, Schreier AB. Persistence of antibodies 25. Al-Robaiy S, Dihazi H, Kacza J, et  al. Metamorphosis of Bor-
to Borrelia burgdorferi in dogs of New York and Connecticut. relia burgdorferi organisms—RNA, lipid and protein composi-
J Am Vet Med Assoc. 1990;196:1064-1068. tion in context with the spirochetes’ shape. J Basic Microbiol.
11. Bowman D, Little SE, Lorentzen L, et al. Prevalence and geographic 2010;50(suppl 1):S5-S17.
distribution of Dirofilaria immitis, Borrelia burgdorferi, Ehrlichia 26. Berger BW, Johnson RC, Kodner C, et al. Cultivation of Borrelia
canis, and Anaplasma phagocytophilum in dogs in the United burgdorferi from human tick bite sites: a guide to the risk of infec-
States: results of a national clinic-based serologic survey. Vet Para- tion. J Am Acad Dermatol. 1995;32:184-187.
sitol. 2009;160:138-148. 27. Hynote ED, Mervine PC, Stricker RB. Clinical evidence for rapid
12. Levine JF, Wilson ML, Spielman A. Mice as reservoirs of the Lyme transmission of Lyme disease following a tickbite. Diagn Microbiol
disease spirochete. Am J Trop Med Hyg. 1985;34:355-360. Infect Dis. 2012;72:188-192.
CHAPTER 51  Lyme Borreliosis 497

28. Antonara S, Ristow L, Coburn J. Adhesion mechanisms of Borrelia 50. Greene CE, Straubinger RK, Levy SA. Borreliosis. In: Greene CE,
burgdorferi. Adv Exp Med Biol. 2011;715:35-49. ed. Infectious Diseases of the Dog and Cat. 4th ed. St Louis, MO:
29. Summers BA, Straubinger AF, Jacobson RH, et al. Histopathologi- Saunders Elsevier; 2012.
cal studies of experimental Lyme disease in the dog. J Comp Pathol. 51. Embers ME, Barthold SW, Borda JT, et al. Persistence of Borrelia
2005;133:1-13. burgdorferi in rhesus macaques following antibiotic treatment of
30. Susta L, Uhl EW, Grosenbaugh DA, et al. Synovial lesions in exper- disseminated infection. PLoS One. 2012;7:e29914.
imental canine Lyme borreliosis. Vet Pathol. 2012;49:453-461. 52. Hodzic E, Feng S, Holden K, et  al. Persistence of Borrelia burg-
31. Levy SA, Duray PH. Complete heart block in a dog seropositive dorferi following antibiotic treatment in mice. Antimicrob Agents
for Borrelia burgdorferi. Similarity to human Lyme carditis. J Vet Chemother. 2008;52:1728-1736.
Intern Med. 1988;2:138-144. 53. Straubinger RK, Straubinger AF, Summers BA, et  al. Status of
32. Kirmizis D, Efstratiadis G, Economidou D, et al. MPGN secondary Borrelia burgdorferi infection after antibiotic treatment and the
to Lyme disease. Am J Kidney Dis. 2004;43:544-551. effects of corticosteroids: an experimental study. J Infect Dis.
33. Dambach DM, Smith CA, Lewis RM, et al. Morphologic, immuno- 2000;181:1069-1081.
histochemical, and ultrastructural characterization of a distinctive 54. Singh SK, Girschick HJ. Molecular survival strategies of the
renal lesion in dogs putatively associated with Borrelia burgdorferi Lyme disease spirochete Borrelia burgdorferi. Lancet Infect Dis.
infection: 49 cases (1987-1992). Vet Pathol. 1997;34:85-96. 2004;4:575-583.
34. Littman MP, Goldstein RE, Labato MA, et al. ACVIM small ani- 55. Hastey CJ, Elsner RA, Barthold SW, et al. Delays and diversions
mal consensus statement on Lyme disease in dogs: diagnosis, treat- mark the development of B cell responses to Borrelia burgdorferi
ment, and prevention. J Vet Intern Med. 2006;20:422-434. infection. J Immunol. 2012;188:5612-5622.
35. Gerber B, Eichenberger S, Wittenbrink MM, et al. Increased preva- 56. Bockenstedt LK, Kang I, Chang C, et  al. CD4+ T helper 1 cells
lence of Borrelia burgdorferi infections in Bernese Mountain Dogs: facilitate regression of murine Lyme carditis. Infect Immun.
a possible breed predisposition. BMC Vet Res. 2007;3:15. 2001;69:5264-5269.
36. Barth C, Straubinger RK, Sauter-Louis C, et al. Prevalence of anti- 57. Poland GA, Jacobson RM. The prevention of Lyme disease with
bodies against Borrelia burgdorferi sensu lato and Anaplasma vaccine. Vaccine. 2001;19:2303-2308.
phagocytophilum and their clinical relevance in dogs in Munich, 58. Poland GA. Vaccines against Lyme disease: what happened and what
Germany. Berl Munch Tierarztl Wochenschr. 2012;125:337-344. lessons can we learn? Clin Infect Dis. 2011;52(suppl 3):s253-s258.
37. Hutton TA, Goldstein RE, Njaa BL, et al. Search for Borrelia burg- 59. Abbott A. Lyme disease: uphill struggle. Nature. 2006;439:524-525.
dorferi in kidneys of dogs with suspected “Lyme nephritis.” J Vet 60. LaFleur RL, Dant JC, Wasmoen TL, et  al. Bacterin that induces
Intern Med. 2008;22:860-865. anti-OspA and anti-OspC borreliacidal antibodies provides a
38. Chou J, Wunschmann A, Hodzic E, et  al. Detection of Borrelia high level of protection against canine Lyme disease. Clin Vaccine
burgdorferi DNA in tissues from dogs with presumptive Lyme bor- Immunol. 2009;16:253-259.
reliosis. J Am Vet Med Assoc. 2006;229:1260-1265. 61. Conlon JA, Mather TN, Tanner P, et  al. Efficacy of a nonadju-
39. Steere AC. Borrelia burgdorferi (Lyme disease, Lyme borrelio- vanted, outer surface protein A, recombinant vaccine in dogs after
sis). In: Mandell GL, Bennett JE, Dolin R, eds. Principles and challenge by ticks naturally infected with Borrelia burgdorferi. Vet
Practice of Infectious Diseases. 7th ed. Philadelphia, PA: Elsevier; Ther. 2000;1:96-107.
2010:3071-3081. 62. Levy SA. Use of a C6 ELISA test to evaluate the efficacy of a whole-
40. Sykes JE. Unpublished observations. 2012. cell bacterin for the prevention of naturally transmitted canine Bor-
41. Liang FT, Jacobson RH, Straubinger RK, et al. Characterization of relia burgdorferi infection. Vet Ther. 2002;3:420-424.
a Borrelia burgdorferi VlsE invariable region useful in canine Lyme 63. LaFleur RL, Callister SM, Dant JC, et  al. One-year duration of
disease serodiagnosis by enzyme-linked immunosorbent assay. immunity induced by vaccination with a canine Lyme disease bac-
J Clin Microbiol. 2000;38:4160-4166. terin. Clin Vaccine Immunol. 2010;17:870-874.
42. Leschnik MW, Kirtz G, Khanakah G, et  al. Humoral immune 64. Nadelman RB, Hanincova K, Mukherjee P, et al. Differentiation of
response in dogs naturally infected with Borrelia burgdorferi sensu relapse from reinfection in recurrent Lyme disease. N Engl J Med.
lato and in dogs after immunization with a Borrelia vaccine. Clin 2012;367:1883-1890.
Vaccine Immunol. 2010;17:828-835. 65. Piesman J, Dolan MC. Protection against Lyme disease spirochete
43. Wagner B, Freer H, Rollins A, et  al. A fluorescent bead-based transmission provided by prompt removal of nymphal Ixodes scap-
multiplex assay for the simultaneous detection of antibodies to ularis (Acari: Ixodidae). J Med Entomol. 2002;39:509-512.
B. burgdorferi outer surface proteins in canine serum. Vet Immunol 66. Nadelman RB, Nowakowski J, Fish D, et  al. Prophylaxis with
Immunopathol. 2011;140:190-198. ­single-dose doxycycline for the prevention of Lyme disease after an
44. Wagner B, Freer H, Rollins A, et al. Antibodies to Borrelia burgdor- Ixodes scapularis tick bite. N Engl J Med. 2001;345:79-84.
feri OspA, OspC, OspF, and C6 antigens as markers for early and 67. Piesman J, Hojgaard A. Protective value of prophylactic antibi-
late infection in dogs. Clin Vaccine Immunol. 2012;19:527-535. otic treatment of tick bite for Lyme disease prevention: an animal
45. Magnarelli LA, Levy SA, Ijdo JW, et al. Reactivity of dog sera to model. Ticks Tick Borne Dis. 2012;3:193-196.
whole-cell or recombinant antigens of Borrelia burgdorferi by ELISA 68. Shapiro ED. Doxycycline for tick bites—not for everyone. N Engl J
and immunoblot analysis. J Med Microbiol. 2001;50:889-895. Med. 2001;345:133-134.
46. Moroff S, Sokolchik I, Woodring T, et al. Classification of Borrelia 69. Centers for Disease Control and Prevention. 2012. http://www.
burgdorferi infection using results of 5 antibody targets in an auto- cdc.gov/lyme/stats/chartstables/casesbyyear.html. Last accessed
mated system [Abstract]. J Vet Intern Med. 2012;26:792. December 30, 2012.
47. Schwaiger M, Peter O, Cassinotti P. Routine diagnosis of Borrelia 70. Bockenstedt LK, Gonzalez DG, Haberman AM, et  al. Spirochete
burgdorferi (sensu lato) infections using a real-time PCR assay. antigens persist near cartilage after murine Lyme borreliosis ther-
Clin Microbiol Infect. 2001;7:461-469. apy. J Clin Invest. 2012;122:2652-2660.
48. Nanagara R, Duray PH, Schumacher Jr HR. Ultrastructural dem- 71. Mullegger RR. Dermatological manifestations of Lyme borreliosis.
onstration of spirochetal antigens in synovial fluid and synovial Eur J Dermatol. 2004;14:296-309.
membrane in chronic Lyme disease: possible factors contributing
to persistence of organisms. Hum Pathol. 1996;27:1025-1034.
49. Chang YF, Novosel V, Chang CF, et al. Experimental induction of
chronic borreliosis in adult dogs exposed to Borrelia burgdorferi–
infected ticks and treated with dexamethasone. Am J Vet Res.
2001;62:1104-1112.
CHAPTER 52

Bartonellosis
Jane E. Sykes and Bruno B. Chomel

Cat fleas (Ctenocephalides felis) play a major role in the


Overview of Bartonellosis in Dogs and Cats transmission of feline Bartonella infections. The presence of cat
First Described: Peru, 1905 (Alberto Barton Thompson) (Bar- fleas is essential for maintenance of the infection within the cat
tonella bacilliformis).1 Identification of Bartonella as the population.4 B. henselae can multiply in the digestive system of
cause of cat scratch disease in humans did not occur until the flea and survive several days in flea feces.5 The main source
1983.2 of infection appears to be flea feces that are inoculated by con-
Causes: The most common species in cats are Bartonella taminated cat claws.6 There is some evidence that Bartonella
henselae and Bartonella clarridgeiae; in dogs, they are Bar- is present in the saliva of cats, but shedding of B. henselae in
tonella vinsonii subsp. berkhoffii and B. henselae. cat saliva has not been clearly documented. Other potential
vectors, such as Pulex flea species, ticks, lice, and biting flies,
Geographic Distribution: Worldwide, with highest preva-
also harbor Bartonella DNA, and experimental transmission of
lence in subtropical and tropical regions
Bartonella birtlesii has been accomplished with Ixodes ricinus
Major Mode of Transmission: Fleas (Ctenocephalides felis); ticks.7 There is epidemiologic evidence that ticks may transmit
possibly other flea species (such as Pulex spp.) and vectors ­Bartonella to dogs.
such as ticks, lice, biting flies Because Bartonella infections are often subclinical in dogs
Major Clinical Signs: Signs that relate to infective endocar- and cats, the full extent to which Bartonella spp. cause disease
ditis (lethargy, fever, cardiac murmur, cough, tachypnea, in naturally infected dogs and cats remains unclear. Bartonella
lameness, neurologic signs) are the most frequent and spp. have been widely studied for their role in a number of
well understood manifestations of bartonellosis in dogs. idiopathic inflammatory disorders of cats, including uveitis,
Bartonella can also cause endocarditis and myocarditis in lymphadenopathy, caudal stomatitis, and rhinitis, without
cats and is suspected to cause (or contribute to) caudal definitive evidence of disease causation. Without doubt, they
stomatitis and possibly other systemic inflammatory dis- are important causes of endocarditis in dogs and occasionally
orders such as uveitis. cause endocarditis and myocarditis in cats (Table 52-3 and
Box 52-1).
Differential Diagnoses: Cats: major differential diagnoses are
other bacterial causes of endocarditis and feline infec- Epidemiology of Bartonella in Cats
tious peritonitis. Dogs: other bacterial causes of endo-
The most common Bartonella species isolated from cats is B.
carditis, other vector-borne diseases such as ehrlichiosis,
henselae. Less commonly, cats are infected with Bartonella clar-
anaplasmosis, borreliosis and babesiosis, and immune-
ridgeiae or Bartonella koehlerae, which also have been linked
mediated inflammatory diseases.
to human disease.5,8 Young cats (≤1 year) are more likely than
Human Health Significance: Bartonella causes cat scratch older cats to be bacteremic, and stray or feral cats are more
disease in immunocompetent humans as well as vasculo- likely to be bacteremic than pet cats.5,9,10 Older cats are more
proliferative disorders in immunocompromised humans. likely to be seropositive than young cats, which likely reflects
the increased chance of exposure over time. The prevalence of
Bartonella bacteremia in cats is also highest in warm, humid cli-
mates (e.g., 68% in the Philippines), whereas it is low in colder
Etiology and Epidemiology climates (e.g., 0% in Norway).5 At least two genotypes of B.
henselae infect cats, type Houston-1 (type I) and type Marseille
Bartonella spp. are fastidious, intraerythrocytic gram-negative (type II). B. henselae type Marseille predominates among cats
bacteria that infect a wide range of domestic and wild mamma- in the western United States, western continental Europe, the
lian host species. Different Bartonella species have adapted to United Kingdom, and Australia; type Houston-1 is dominant
specific mammalian reservoir hosts and can infect and occasion- in Asia (Japan and the Philippines).5 However, within a given
ally cause disease in alternative, incidental hosts. More than 10 country, the prevalence of these types varies among cat popula-
species of Bartonella infect cats or dogs worldwide (Tables 52-1 tions, and type Houston-1 is more often isolated from humans
and 52-2). Bartonella infections are important because cats are with bartonellosis, even when type Marseille is more prevalent
the principal reservoir host for Bartonella henselae, the main in the cat population. Some molecular typing methods reveal an
cause of cat scratch disease (CSD) in humans, and subclinical even broader genetic diversity among feline strains, and most
bacteremia in cats is widespread (8% to 56% of healthy cats of the strains that infect humans cluster in a limited number of
worldwide).3 groups.

498
CHAPTER 52  Bartonellosis 499

TABLE 52-1 TABLE 52-3


Species of Bartonella Known to Infect Cats Examples of Clinical Manifestations That May, in Some
Circumstances, Be Associated with Bartonella Infection in
Bartonella Primary Primary Naturally Infected Cats
Species Reservoir Vector
B. henselae Cats Cat flea (Ctenocephalides Strength of Clinical Bartonella
felis) Association Manifestation Species
B. clarridgeiae Cats Cat flea Definite Endocarditis26 B. henselae
B. koehlerae Probably cats Cat flea Myocarditis and
B. quintana Human Human body louse ­diaphragmatic myositis27
­(Pediculus humanus) Probable Osteomyelitis28 B. v. berkhoffii
B. bovis Ruminants Biting flies? Systemic reactive
(cattle, deer) ­angioendotheliomatosis29
B. vinsonii Coyotes, do- Unknown (fleas, ticks?) Possible* Caudal stomatitis32-37 Unknown
subspecies mestic dogs Lower urinary tract
berkhoffii ­disease32,36
Uveitis30,31
Lymphadenopathy37
TABLE 52-2 Fever38
Species of Bartonella Known to Infect Dogs *Results of experimental studies are conflicting or there is insufficient
evidence to prove an association with disease.
Bartonella Species Primary Reservoir Primary Vector
B. henselae Domestic cats Cat flea (Ctenoce-
phalides felis) Epidemiology of Bartonella in Dogs
B. vinsonii subsp. Coyotes, domestic Unknown (fleas, Dogs are most commonly infected with Bartonella vinsonii
berkhoffii dogs, foxes ticks?) subsp. berkhoffii or B. henselae. Domestic and wild dogs are
B. rochalimae Wild carnivores, Fleas (Pulex thought to be the natural reservoir for B. v. berkhoffii, because
domestic dogs ­irritans) B. v. berkhoffii establishes prolonged bacteremia in dogs. In
California, 35% of coyotes (Canis latrans) tested were seroposi-
B. clarridgeiae Domestic cats Cat flea tive, and 28% of coyotes from a highly endemic region were
B. koehlerae Domestic cats Unknown bacteremic.5 Other Bartonella species have also been detected in
B. quintana Human Body louse (Pedic- dogs (see Table 52-2). Novel species also have been identified
ulus humanus) in dogs from Thailand, Sri Lanka, and the Mediterranean.11-13
(in humans) Because it is often difficult to culture Bartonella from domes-
tic dog blood, prevalence studies in dogs are usually based on
B. washoensis California ground Unknown
antibody detection. Unfortunately, this correlates poorly with
squirrel
bacteremia. In addition, serologic cross-reactivity occurs among
B. bovis Ruminants (cattle, Unknown Bartonella species so seroprevalence studies cannot be Barton-
deer) ella species-specific. The prevalence of antibodies to B. v. berk-
B. elizabethae Rats Oriental rat flea hoffii in dogs is highest in tropical or subtropical regions. For
(Xenopsylla example, seroprevalences as high as 47% were detected in stray
cheopis) dogs from Morocco.14 In the southeastern United States, the
B. grahamii Wild mice Rodent fleas prevalence of B. henselae antibodies in healthy dogs was 10%;
(Ctenophthal- a higher prevalence (27%) was found in sick dogs.15 Risk fac-
mus nobilis) tors identified in dogs in the southeastern United States were
tick exposure, residence in a rural environment, roaming, and
B. taylorii Wild mice Rodent fleas
outdoor exposure.16 The prevalence of B. henselae antibodies
(Ctenophthal-
in sick dogs from the western United States is less than 2%.17
mus nobilis)
B. vinsonii subsp. White-footed Unknown Clinical Features
arupensis mouse
B. volans–like Southern flying Unknown Signs and Their Pathogenesis
squirrel After infection, Bartonella replicates in erythrocytes and can also
“Candidatus Dogs, jackals Unknown infect endothelial cells and bone marrow progenitor cells.18 It
B. merieuxii” then establishes chronic, often subclinical bacteremia, which can
(strain HMD) last for weeks, months, or even more than a year and may be a
specific adaptation to a mode of transmission by blood-sucking
500 SECTION 2  Bacterial Diseases

BOX 52-1
Descriptive Case Reports or Small Case Series Where Bartonella Has Been Detected within Lesions in Dogs using Culture or PCR

Endocarditis Systemic granulomatous disease and


B. v. berkhoffii45-47 sialometaplasia
B. henselae45,48 B. henselae
B. clarridgeiae46 B. v. berkhoffii55
B. koehlerae48
B. quintana49 Chronic erosive polyarthritis
B. rochalimae50 B. henselae
B. v. berkhoffii56
Chronic lymphocytic hepatitis with copper
accumulation (Doberman) Massive post-traumatic seroma
B. clarridgeiae51 B. henselae
B. v. berkhoffii57
Systemic pyogranulomatous disease, hyperviscosity
syndrome Hemangiopericytoma
Bartonella spp.52 B. v. berkhoffii or B. henselae29

Pyogranulomatous lymphadenitis Epistaxis


B. henselae53 B. v. berkhoffii or B. henselae58

Granulomatous hepatitis Meningoradiculoneuritis and pyogranulomatous


B. henselae51 dermatitis or panniculitis
B. v. berkhoffii59
Peliosis hepatis
B. henselae54 Bacillary angiomatosis
B. v. berkhoffii60

arthropods.19 Infection of endothelial cells may be more likely Clinical Manifestations in Cats
to occur in incidental hosts and appears to be necessary for the Experimental infections of specific pathogen free cats with
development of disorders such as vasculoproliferative disease and Bartonella have shed light on disease processes that can fol-
endocarditis. Bartonella is thought to cause vasculoproliferative low infection of cats with Bartonella. Cats infected with
disease via cytokine-induced stimulation of endothelial cell pro- B. henselae develop a small papule at the site of inoculation,
liferation and inhibition of endothelial cell apoptosis. Virulence and transient fever and lymphadenopathy.21-24 Transient neu-
factors characterized in Bartonella species include adhesins, heme rologic signs (such as staring and behavioral abnormalities) and
acquisition mechanisms, type IV secretion systems, and a low- reproductive disorders can also occur. Some cats infected with
potency lipopolysaccharide, among others.20 Host immunocom- B. henselae and/or B. clarridgeiae show no clinical signs, and
promise, such as due to genetic susceptibility (e.g., breed-related gross lesions are lacking at necropsy. However, histopathology
immunodeficiency syndromes), poor nutrition, overcrowding, reveals peripheral lymph node hyperplasia, splenic follicular
co-infections with other pathogens, immunosuppressive drug hyperplasia, lymphocytic cholangitis/pericholangitis, lympho-
treatment, or defects in normal host barriers (such as congenital cytic hepatitis, lymphoplasmacytic myocarditis, and/or intersti-
valvular disease in dogs with endocarditis), may also influence tial lymphocytic nephritis.21,23
whether disease ultimately develops in infected dogs and cats. In naturally infected cats, Bartonella can rarely cause endo-
The vast majority of infected dogs and cats show no overt carditis and myocarditis. Generally speaking, infective endocar-
clinical signs of illness when bacteremic. In some situations, dis- ditis is rare in cats, but a few of the reported cases have been
ease manifestations that occur in Bartonella spp.–infected dogs associated with Bartonella infection. B. henselae DNA has been
and cats are otherwise uncommon to rare and/or known to be amplified from aortic valvular vegetative lesions of cats, with
associated with Bartonella infection in human patients; organ- visualization of the organism in lesions using silver stains.25,26
isms may also have been visualized with silver stains using light Dramatic pyogranulomatous myocarditis and diaphragmatic
microscopy in affected tissues (e.g., endocarditis or myocarditis myositis were associated with B. henselae infection in two
lesions). In other situations, nonspecific clinical signs are pres- cats that died in a North Carolina shelter, and organisms were
ent (e.g., uveitis, gingivostomatitis, chylothorax, polyarthritis, detected in lesions with silver stains and immunohistochemis-
chronic rhinitis, idiopathic lower urinary tract disease, lymph- try (IHC) (Figure 52-1).27 Carpal and metacarpal osteomyelitis
adenopathy, reproductive disease, or neurologic disorders), and was also associated with a B. v. berkhoffii infection in a cat;
it has been impossible to clearly assign a role to Bartonella in Bartonella DNA was detected in bone lesions as well as in the
disease causation. A clear understanding of the role of Barton- blood.28 The DNA of B. v. berkhoffii and B. henselae was also
ella in disease causation has also been thwarted by limitations of detected in cardiac tissues of several cats with systemic reactive
diagnostic tests available for detection of infection. angioendotheliomatosis.29
CHAPTER 52  Bartonellosis 501

B C
FIGURE 52-1  A, Granulomatous myocarditis associated with Bartonella henselae infection in an 8-week-old female domestic shorthair introduced to a shelter that was ridden
with fleas. B, Histopathology of the heart of the affected cat showing intralesional agyrophilic bacteria (arrows). Warthin-Starry stain. C, Short bacilli (arrows) in an inflammatory focus
are immunoreactive (brown) for Bartonella henselae–specific monoclonal antibody. Immunohistochemistry with diaminobenzidine chromogen, hematoxylin counterstain. (A, Courtesy
Drs. Jack Broadhurst and Edward Breitschwerdt. From Varanat M, Broadhurst J, Linder KE, et al. Identification of Bartonella henselae in 2 cats with pyogranulomatous myocarditis and
diaphragmatic myositis. Vet Pathol 2012;49:608-611.)

Bartonella has been investigated for its role in a large number of dogs with endocarditis. The most common species identi-
of idiopathic conditions in cats (see Table 52-3).26-38 These include fied has been B. v. berkhoffii. In dogs, Bartonella endocarditis
uveitis,30-32 caudal gingivostomatitis,32-37 fever of unknown usually involves the aortic valve, although the mitral valve is
origin,38 lower urinary tract disease,32,36,39 chronic kidney dis- occasionally affected, so the possibility of bartonellosis should
ease,32,36 pancreatitis,40 lymphadenopathy (Figure 52-2),37 neu- not be ruled out in dogs with mitral valve endocarditis. Dogs
rologic disease,32,41,42 pododermatitis,43 and chronic idiopathic with Bartonella endocarditis are more likely to be afebrile
rhinosinusitis.44 There is currently no convincing and repeatable and more likely to have congestive heart failure than dogs
scientific evidence that Bartonella infection causes any of these with other causes of endocarditis, and their median survival
conditions. Studies have varied in the methodology used to detect time is shorter (Figure 52-3).61 Complications of Bartonella
infection (serology, culture, and/or PCR assays), and many of the endocarditis include thromboembolic disease and neutrophilic
conditions investigated likely have multifactorial etiologies. An polyarthritis.
understanding of the significance of Bartonella in these conditions Bartonella, or Bartonella DNA, has been detected in lesions
will probably require prospective study of large numbers of cats from dogs with a variety of chronic pyogranulomatous or gran-
with clearly defined clinical illness using Bartonella culture and/ ulomatous inflammatory and vasculoproliferative diseases (see
or PCR assays. Box 52-1). Bartonella was detected in a dog with peliosis hepa-
tis and a dog with bacillary angiomatosis, rare conditions that
Clinical Manifestations in Dogs are strongly associated with Bartonella infection in humans.54,60
Bartonella spp. are important causes of blood-culture–negative Peliosis hepatis and bacillary angiomatosis are vasculoprolifera-
endocarditis in dogs (see Box 52-1).25,29,45-60 Infection with tive diseases characterized by the presence of blood-filled pro-
Bartonella was identified in 6 (19%) of 31 dogs with culture– liferations of vascular tissue in the liver and skin, respectively.
negative endocarditis in California.61 Worldwide, the DNA of Based on case reports and seroprevalence studies, Bartonella is
several Bartonella species has been detected in the heart valves suspected to cause polyarthritis, epistaxis, thrombocytopenia,
502 SECTION 2  Bacterial Diseases

and/or splenomegaly in dogs.17 However, these associations


could also reflect the presence of other detected or undetected,
co-transmitted vector-borne pathogens that cause these dis-
orders. Bartonella has been investigated for its role in canine
lymphoma62; neurologic disease63; splenic disorders including
lymphoid nodular hyperplasia, hemangiosarcoma, and fibrohis-
tiocytic nodules64; idiopathic cavitary effusion65; and idiopathic
rhinitis.66 As for cats, the role that Bartonella plays in these
conditions, if any, is unclear and requires further study.

Physical Examination Findings


In dogs with Bartonella endocarditis, physical examination
findings include lethargy, inappetence and thin body condition;
lameness or recumbency due to thromboembolic complica-
tions, weakness, or polyarthritis; joint effusion; and, uncom-
monly, fever (as high as 104.9°F [41°C]). Evidence of cardiac
disease and congestive heart failure is often present, including
respiratory distress, cough, increased lung sounds, and sys-
tolic and/or diastolic heart murmurs, cardiac arrhythmias, and
pulse deficits. Neurologic signs such as anisocoria and obtun-
dation can occur in dogs with thromboembolism that involves
the central nervous system. There may also be evidence of flea
FIGURE 52-2  Lymph node of a 1-year-old, retrovirus-negative, flea-ridden cat with a or tick infestations in some dogs. Other physical examination
3-month history of fever, mild lethargy, and generalized peripheral lymphadenopathy but findings that might be present in dogs with other manifesta-
a normal appetite. The right axillary and both mandibular lymph nodes measured 3 cm in
tions of bartonellosis are mass lesions that involve the nasal
diameter; the remaining peripheral lymph nodes were 2 cm in diameter. A serum chemis-
try profile revealed hyperglobulinemia (6.5 g/dL). Serum protein electrophoresis revealed cavity, lymph nodes, or salivary glands; splenomegaly or hepa-
a polyclonal gammopathy. Histopathology of the right axillary, right mandibular and right tomegaly; cavitary effusions; peripheral edema; epistaxis; or
popliteal lymph node showed lymphofollicular hyperplasia and marked neutrophilic, lym- nodular skin lesions.
phocytic, and histiocytic capsulitis and perilymphadenitis. Note the florid capsulitis to the Physical examination findings in cats with Bartonella endo-
right of the arrows, which indicate the outer extent of the lymph node parenchyma. Silver carditis or myocarditis have included fever, lethargy, respiratory
stains (Warthin-Starry and Steiner stains) showed a small amount of granular staining distress, cardiac murmurs, and arrhythmias.26,27
within cells, which possibly represented intracellular agyrophilic bacteria. The DNA of Bar-
tonella henselae and Bartonella clarridgeiae was detected in the lymph node using PCR, Diagnosis
and blood cultures were positive for B. henselae. Interestingly, the cat was seronegative to
B. henselae and B. clarridgeiae.
Diagnosis of bartonellosis is based on the presence of consistent
clinical abnormalities (especially endocarditis or myocarditis,
but also other systemic inflammatory or vasculoproliferative
diseases), histopathologic findings, and positive culture or PCR

100

90

80

70 Dogs with IE caused by Bartonella spp. infection


Dogs with IE caused by other organisms
Percent survival

60

50

40

30

20

10

0
0 250 500 750 1000 1250 1500 1750 2000
Time (days)
FIGURE 52-3  Kaplan-Meier survival curve comparing survival of dogs with Bartonella infectious endocarditis (IE) with that of dogs with IE caused by other pathogens. (From Sykes
JE, Kittleson MD, Pesavento PA, et al. Evaluation of the relationship between causative organisms and clinical characteristics of infective endocarditis in dogs: 71 cases (1992-2005). J Am
Vet Med Assoc 2006;228:1723-1734.)
CHAPTER 52  Bartonellosis 503

TABLE 52-4
Diagnostic Assays Available for Bartonella Infection in Dogs and Cats
Assay Specimen Type Target Performance
Culture Whole blood, tissue speci- Bartonella spp. Confirms infection and allows species identification. Special-
mens obtained at nec- ized culture conditions required; best performed in labo-
ropsy (e.g., heart valve, ratories with special expertise. Requires several weeks’
myocardium) or by incubation. Sensitivity is especially low in dogs because
biopsy (e.g., liver biopsy, of low-level or intermittent bacteremia. Use of BAPGM
skin biopsy) enrichment media followed by PCR may increase sensitiv-
ity but may increase the chance of false-positive results
due to PCR-related contamination. Positive culture results
alone do not apply disease causation, because healthy
dogs and cats can have Bartonella in their blood.
Histopathology with Tissue specimens obtained Bartonella spp. Low sensitivity, but supports disease causation when
organism ­detecting by biopsy or necropsy organisms positive and properly performed and interpreted.
using silver stains
or IHC
Serology Serum Antibodies to Variable cross-reactivity between Bartonella species;
Bartonella spp. species-specific assays are required. Immunofluorescent
antibody, ELISA, and Western immunoblot assays are
available. Results correlate poorly with bacteremia.
Positive results do not imply disease causation, because
a significant proportion of healthy dogs and cats are
seropositive. Positive titers are usually present in dogs
and cats with endocarditis.
PCR Whole blood, splenic or Bartonella spp. Confirms active infection more rapidly than culture.
lymph node aspirates, DNA Sensitivity and specificity may vary depending on assay
tissue specimens obtained design and specimen type; may not necessarily be more
at necropsy or biopsy sensitive than culture. Because healthy animals may be
PCR positive, positive PCR results must be interpreted
in light of the clinical signs. False positive results may
occur as a result of PCR-related contamination.

assay results on blood and affected tissues (Table 52-4). It may numbers of circulating band neutrophils, lymphocytosis, and/
be impossible to identify Bartonella as the cause of disease if or monocytosis are present.49,68,69 The biochemistry panel
the organism is detected only in the blood and if affected tis- may reveal mild azotemia (creatinine <3 mg/dL), mild hypoal-
sues (e.g., valvular vegetations) cannot be obtained for analy- buminemia, and, less often, hyperglobulinemia. Isosthenuria,
sis, because healthy dogs and especially cats can be bacteremic. proteinuria, cylindruria, and slightly increased urine protein-to-
Other possible causes of disease must also be investigated, and creatinine ratios have been present in the urine of some affected
a response to antibiotic treatment could be evaluated (see Case dogs. Dogs with hepatic disorders associated with Bartonella
Example).67 It is even more difficult to establish a definitive infection have had moderately increased activities of serum ALT
diagnosis based on serology, but when culture and PCR assay and/or ALP.51,54
results are negative, positive serologic test results in dogs may
be of some value in the presence of consistent clinical abnor- Diagnostic Imaging
malities. More specifically (and as occurs in humans), dogs with Plain Radiography
Bartonella endocarditis usually have high antibody titers to Thoracic radiographs in dogs with Bartonella endocarditis
Bartonella spp. Organism detection tests are insensitive in dogs often show evidence of congestive heart failure, with venous
because dogs tend to have very low levels of bacteremia when engorgement and pulmonary edema (Figure 52-4). Lung lobe
compared with cats. consolidation and hilar lymphadenopathy were described in a
dog with systemic granulomatous disease and sialometaplasia.55
Laboratory Abnormalities
Hematologic and biochemistry abnormalities in dogs and cats Abdominal Ultrasound
with Bartonella endocarditis are nonspecific and often mild. Ultrasound of dogs with systemic granulomatous or pyogranu-
The CBC may show a mild nonregenerative anemia (31% to lomatous disease may reveal lymphadenomegaly and organo-
39%), variable leukocytosis primarily due to neutrophilia (up megaly,52 and mixed echogenic patterns within the liver.51 The
to 43,000 neutrophils/µL), and mild thrombocytopenia (usually dog with peliosis hepatis had ascites and multiple, small nodular
above 100,000 platelets/µL). In a few dogs, low to moderate masses and hypoechoic cystic lesions within the liver.54
504 SECTION 2  Bacterial Diseases

A
A

B
B
FIGURE 52-5  Echocardiogram images from a 13-year old German shepherd dog
FIGURE 52-4  Lateral thoracic radiograph from a 3-year-old female spayed Dober- with blood culture–negative endocarditis. The dog also had biopsy-confirmed glomerulo-
man pinscher with early congestive heart failure due to endocarditis, before (A) and after nephritis. IFA serology for Bartonella vinsonii subsp. berkhoffii, Bartonella clarridgeiae, and
(B) treatment with furosemide. The dog was seroreactive to Bartonella vinsonii subsp. Bartonella henselae were positive at titers of 1:512, 1:1024, and 1:128, respectively. On the
berkhoffii with a titer of 1:256 and Bartonella henselae with a titer of 1:64. A, There is mild long-axis view (A) there is an irregular hyperechoic lesion attached to the aortic valve
cardiomegaly with left atrial enlargement. There is increased pulmonary interstitial opac- cusps (arrow), and the cusps appear thickened. On the short-axis view (B), the normal
ity that is most pronounced in the accessory lung lobe, as well as cranial to the heart.  “Mercedes Benz” sign has been destroyed by the aortic valvular thickenings.
B, Pulmonary infiltrates have significantly resolved.

tubes, and then chilled or frozen during shipment. Collection


Echocardiography of blood into plastic EDTA tubes allows the tubes to be fro-
Echocardiography in dogs and cats with Bartonella endocar- zen directly, which releases organisms from erythrocytes after
ditis reveals oscillating valvular vegetations or thickened and which they are plated onto special media. Blood should be
hyperechoic valve leaflets, which usually involve the aortic valve sent to laboratories familiar with the culture of these fastidious
(Figure 52-5). There may be evidence of aortic regurgitation or organisms, and the laboratories should be contacted for specific
insufficiency and eccentric hypertrophy of the left ventricle in instructions regarding specimen collection and submission. Spe-
some dogs. cial media (such as fresh chocolate agar, or brain-heart infusion
agar enriched with 5% to 10% blood) and culture conditions
Microbiologic Tests (5% CO2, temperatures of 35°C to 37°C) are used to enhance
Culture the likelihood of isolation of Bartonella.62 Extended incubation
Culture of blood or other tissues for Bartonella allows defini- times of up to 6 to 8 weeks are often required. Serial Bartonella
tive diagnosis of infection. However, blood culture requires spe- blood cultures may enhance the chance of a positive result. The
cial techniques and can be insensitive because of the relapsing use of an insect growth medium (Bartonella Alpha Proteobacte-
nature of bacteremia. In contrast to the situation in cats, culture ria Growth Medium, or BAPGM) that is enriched with multiple
has especially low sensitivity in dogs because of the low level micronutrients and defibrinated sheep blood for pre-enrichment
of bacteremia in dogs (especially when they act as incidental culture of blood and potentially other tissues, followed by PCR
hosts and develop disease). Blood for culture should be obtained assay of the liquid culture and subinoculation of the liquid cul-
using sterile technique and the blood placed in EDTA-containing ture onto agar plates, may enhance the sensitivity of culture for
CHAPTER 52  Bartonellosis 505

diagnosis of Bartonella infections of dogs. It also has the poten-


tial to yield other fastidious organisms not cultured under rou-
tine conditions that may play a role in endocarditis.70

Molecular Diagnosis Using the Polymerase Chain Reaction


A variety of PCR-based assays have been developed for rapid
detection of Bartonella DNA in blood or other tissues. Some
veterinary diagnostic laboratories offer real-time PCR assays
for detection of Bartonella species as a component of vector-
borne PCR panels. Some assays detect all species of Bartonella,
whereas others are species-specific. Genetic analysis of the resul-
tant PCR products can also be performed to identify the infect-
ing species. The clinical performance of many of these assays
when compared with culture has not been well studied. When
used directly on blood specimens, PCR assays may be no more
sensitive than culture for detection of Bartonella, and detection
of DNA does not always equate to detection of living organ-
isms. False-positive PCR assay results due to laboratory con- A
tamination have the potential to occur.

Serologic Diagnosis
Immunofluorescent antibody (IFA), ELISA, and Western immu-
noblot assays are available for detection of antibodies to Bartonella
species. Some degree of cross-reactivity occurs among Bartonella
species, but multiple species-specific assays still should be per-
formed to increase the chance of antibody detection. Paired titers
are not valuable because disease typically results from chronic and
persistent infection. If serologic testing is performed in dogs, at a
minimum it should include testing for B. henselae, B. v. berkhoffii,
B. rochalimae, and B. clarridgeiae. Measurement of serum anti-
bodies to Bartonella has a limited ability to help determine whether
a sick animal has an active Bartonella infection. In both cats and
dogs, bacteremia can occur in the absence of detectable antibodies
(see Figure 52-2), and seropositive cats and dogs are not always
bacteremic. Fewer than half of antibody-positive cats are bactere-
mic, and 3% to 15% of antibody-negative cats are bacteremic.67
Because of this, serology is not recommended for routine diagnosis
of Bartonella infection in cats. Because of the low sensitivity of
PCR and culture in dogs, serology could be considered when these B
assays and routine blood cultures are negative (or in conjunction FIGURE 52-6  Gross pathologic findings (A) and histopathology (B) of the aortic
with PCR and culture) to support a diagnosis of Bartonella endo- valve from a dog with Bartonella endocarditis. A, The periphery of the valve is eroded. Vari-
carditis (which is usually associated with high antibody titers). ably sized, mineralized nodules expand the valve leaflet. There are multifocal, slightly raised,
dull white plaques on the surface of the adjacent aorta and ventricular endocardium. B, Aor-
Pathologic Findings tic valve, aorta, and ventricular septum. The distal leaflet is fragmented and contains mineral 
Gross necropsy of dogs and cats with Bartonella endocarditis deposits (arrow) and hemorrhage. At the base of the valve, the endothelial surface is lifted
and there are broad bands of loose connective tissue, small vessels, and endothelial-lined
may reveal evidence of congestive heart failure, with lung con-
clefts (arrowheads). H&E stain. Bar = 180 µm. (Reprinted from Pesavento PA, Chomel BB, Kas-
gestion and edema. Single or multiple, variably sized nodules are ten RW, et al. Pathology of Bartonella endocarditis in six dogs. Vet Pathol 2005;3:370-373.)
typically present on the periphery of valvular leaflets, which may
be small and firm or large, granular, and friable (Figure 52-6).
White plaques may be found adherent to the adjacent endo- scattered through other parts of the liver. Nodular granuloma-
cardial surfaces. Histopathology reveals chronic inflammation, tous or pyogranulomatous lesions in lymphoid or other tissues
with fibrous tissue and extensive mineral deposits. The presence (such as the liver, lymph nodes, and nasal cavity) have also been
of valvular mineralization should raise suspicion for Bartonella described in dogs with bartonellosis,47,51,71 and nodular pyo-
endocarditis, because mineralization is uncommonly associated granulomatous inflammatory lesions with intralesional bacteria
with endocarditis caused by other bacteria.61 Small bacilli may were described in the myocardium and diaphragm of cats with
be seen within lesions with silver stains or immunohistochemical Bartonella myocarditis and diaphragmatic myositis.27
stains for B. henselae.26,27,60 Silver stains must be interpreted with
caution, because necrotic debris and nuclear material can mimic Treatment and Prognosis
positive staining. Gross lesions in the dog with peliosis hepatis
consisted of multiple, 0.1- to 0.3-cm cysts in the liver that were Efficacy has not been established for any antimicrobial that could
filled with cloudy, serosanguineous fluid.54 The cysts were lined be used to eliminate Bartonella bacteremia in cats or dogs. In
with endothelial cells, and there was evidence of telangiectasia humans, doxycycline, erythromycin, and rifampin are the most
506 SECTION 2  Bacterial Diseases

TABLE 52-5
Medications That Could Be Used to treat Bartonella spp.
Infections in Sick Dogs and Cats Suspected to have Bartonellosis
Dose Interval
Drug (mg/kg) Route (hours)
Ampicillin 10-20 IV 6-8
sodium
Gentamicin 9-14 (dogs), IV, IM, 24
sulfate* 5-8 (cats) SC
Amoxicillin- 12.5-25 PO 8
clavulanate
Doxycycline 10 PO 12
Rifampin† 5 PO 12
Azithromycin 5-10 PO 24
Marbofloxacin 2.75-5.5 PO 24
Enrofloxacin‡ 5 (cats), PO 24
5-20 (dogs)

*Use in combination with ampicillin for initial treatment of endocardi-


tis; other aminoglycosides could be substituted for gentamicin (such as
amikacin). Monitor renal values during treatment.
†Use in combination with doxycycline; avoid monotherapy with ri-

fampin because of the potential for rapid selection for resistant bacteria.
May cause gastrointestinal adverse effects. FIGURE 52-7  A crop of cutaneous bacillary angiomatosis lesions on the elbow of an
‡Other fluoroquinolones are preferred in cats due to the higher risk of
AIDS patient. All lesions involuted with doxycycline treatment. (From Slater LN, Welch DF.
retinal toxicity with enrofloxacin. Doses of 5 mg/kg q24h should never Bartonella, including cat-scratch disease. In: Mandell GL, Bennett JE, Dolin R, eds. Mandell,
be exceeded in cats. Douglas and Bennett’s Principles and Practice of Infectious Diseases, 7 ed. Philadelphia, PA:
Churchill Livingstone Elsevier; 2010:2995-3009.)
frequently recommended antimicrobials for treatment of Barton-
ella infection, but clinical improvement has been reported after a persistent intracellular infection. Dogs and cats can be co-
the use of penicillin, gentamicin, ceftriaxone, ciprofloxacin, or infected with multiple different Bartonella species and geno-
azithromycin. Treatment for 2 weeks is recommended for immu- types. Vaccination and challenge with the same or different
nocompetent patients (e.g., for CSD), and for a minimum of 6 Bartonella species results in variable protection, and protection
weeks in immunocompromised humans. Relapses in bacteremia, is generally species specific.74-76
fever, or other disease manifestations have been reported in immu-
nocompromised humans, despite a 6-week treatment regimen. Prevention
The prognosis for Bartonella endocarditis in dogs and cats is
generally poor. Supportive care and medications such as furo- Prevention of Bartonella infections in cats and dogs is best
semide to manage congestive heart failure are often required. A accomplished by avoiding exposure to arthropod vectors. Flea
combination of a β-lactam and an aminoglycoside or doxycy- control measures, such as use of ectoparasiticides, can interrupt
cline is recommended for treatment of Bartonella endocarditis transmission of B. henselae among cats and should be main-
in human patients, and valve replacement is usually indicated tained year-round.67,77 Housing cats indoors may also reduce
(see Chapter 86).72 Oral antimicrobial alternatives for dogs that exposure to fleas. B. henselae and B. clarridgeiae have been
are stable include high-dose doxycycline, alone or in combina- transmitted through inoculation of infected cat blood; therefore,
tion with rifampin; azithromycin; or fluoroquinolones, alone or cats of unknown Bartonella status or cats that are positive by
in combination with amoxicillin (Table 52-5). Resistance of B. culture, serology, and/or PCR for Bartonella should be excluded
henselae to azithromycin and fluoroquinolones has been docu- as blood donor cats.
mented.73 Data from experimentally or naturally infected cats
indicate that a high dose of doxycycline may be necessary to elim- Public Health Aspects
inate Bartonella infection in cats. Amoxicillin–clavulanic acid
may also be an appropriate choice for treatment of infected cats Cats are the major reservoirs for B. henselae, the principal
when a definitive diagnosis is not known. If there is no response cause of CSD in immunocompetent humans. CSD is char-
after 7 days, a change to a fluoroquinolone or azithromycin could acterized by formation of a pustule at the site of inoculation
be considered.67 Extreme care should be taken to avoid scratches (a cat scratch) within 14 days, followed by development of local
or bites when administering drugs to bacteremic cats. lymphadenopathy and malaise 1 to 3 weeks after the scratch (or
bite), which can persist for weeks to months. Most cases of CSD
Immunity and Vaccination are self-limiting after several months and respond minimally to
antimicrobial drug treatment. Occasionally CSD is complicated
There are no vaccines for prevention of bartonellosis in cats or by other disorders such as encephalopathy, relapsing bactere-
dogs. Bartonella evades the host immune system and establishes mia, osteomyelitis, neuroretinitis, or endocarditis, which require
CHAPTER 52  Bartonellosis 507

antimicrobial drug treatment. Vasculoproliferative diseases such


as bacillary peliosis (e.g., peliosis hepatis) and bacillary angioma- BOX 52-2
tosis (Figure 52-7) are among the most common complications
in immunocompromised humans.20 Young cats are most often
Advantages and Disadvantages of Routine Testing
implicated in transmission to humans, because young cats are of Healthy Cats for Bartonella spp. Infection
more likely to be bacteremic than older cats. Dogs are less likely
to act as reservoirs for human disease when compared with cats, Advantages
because the prevalence of Bartonella infection and the magni- Cats with positive Bartonella test results can be eliminated
tude of bacteremia in dogs are lower than that in cats. Neverthe- as blood donors or breeding animals
less, dogs have occasionally been implicated in transmission of Cats with negative Bartonella test results (serology and
Bartonella to humans through bite wounds or scratches, and culture or PCR) may be safer pets than cats that test
to date, all Bartonella species identified in sick dogs are also positive
pathogenic or potentially pathogenic in humans. Immunocom- Testing cats for Bartonella spp. may allow veterinarians to
promised humans should preferably choose adult, well-social- avoid claims or litigation
ized cats over kittens as pets and should be properly educated
in regard to the need for flea prevention. Cats’ claws should be Disadvantages
trimmed regularly, but there is no evidence that declawing pre- Bartonella serology and PCR can be falsely positive
vents transmission. Animal scratches or bites should be avoided, Some cats with positive serologic test results may have
and if they occur, they should be washed thoroughly with soap eliminated infection and be immune to reinfection
and water, after which prompt medical attention should be Cats with negative test results may still be bacteremic, or
sought. The Guidelines for Preventing Opportunistic Infections become bacteremic within the near future
among HIV-Infected Persons (United States Public Health Ser- Detection of positive Bartonella spp. test results in some
vice and Infectious Diseases Society of America) state “No evi- situations may lead to needless euthanasia
dence indicates any benefits to cats or their owners from routine A focus on testing may lead to underappreciation of the
culture or serologic testing of the pet for Bartonella infection.”78 need for other relevant health care such as flea control
There is also no antimicrobial drug treatment that reliably elimi- There is no treatment that reliably eliminates bacteremia
nates bacteremia from cats, and administration of antimicrobial in healthy cats, and antimicrobial drug treatment of cats
drugs to cats may result in unnecessary bites, scratches, and con- has not been shown to prevent human infection
tact with cat saliva. Pros and cons for the testing of healthy cats Treatment of positive cats may increase the risk of human
for Bartonella infection are shown in Box 52-2.67 infection due to an increased risk of bites and scratches
Bartonella has the potential to be transmitted to veterinar- Treatment of positive cats may lead to unnecessary life-
ians as a result of scratches or bite wounds as well as needle- threatening adverse drug reactions such as esophageal
stick injuries.79,80 Because of the high prevalence of Bartonella strictures (doxycycline) or blindness (fluoroquinolones);
bacteremia in stray and young cats, veterinarians should take it also selects for antimicrobial-resistant bacteria
care to avoid these injuries (consider use of sedation whenever
possible if necessary), thoroughly wash wounds that occur,
and seek medical attention early if necessary. Gloves should be
worn if skin contact with animal blood or saliva is anticipated. possibly through blood transfusions.81 Human body lice pri-
Although cats and dogs are important reservoirs of Bar- marily transmit B. quintana among humans (especially the
tonella, humans can also develop bartonellosis through other homeless), which causes relapsing fever (trench fever) and
routes of transmission, such as via other arthropods and endocarditis.

CASE EXAMPLE been housed indoors ever since. She was neutered at 8
months of age without complication. Five months earlier,
she traveled briefly to Oregon with the owner. She lived with
Signalment: “Pesto,” a 16-month-old female spayed domestic a dog and a retrovirus-negative kitten that was introduced
medium hair cat from Rio Vista in northern California 5 months ago. One other sibling had died of FIP, which was
History: Pesto was evaluated at the University of California, confirmed on necropsy. Pesto was up to date on vaccinations
Davis, Veterinary Medical Teaching Hospital for progressive for respiratory viruses, panleukopenia virus, and rabies. She
lethargy over the previous month. Since obtained as a kitten, was not receiving any medications.
she had shown intermittent signs of upper respiratory tract Physical Examination:
disease, characterized by sneezing, serous to mucopurulent Body Weight: 2.9 kg.
nasal and ocular discharges, inappetence, blepharospasm, General: Quiet, alert, responsive, and hydrated. T = 103°F
tachypnea, and a cough, for which she had been treated (39.5°C), HR = 240 beats/min, RR = 40 breaths/minute, moist
with amoxicillin or doxycycline on at least three separate and pink mucous membranes, CRT <2 s.
occasions. Between episodes she appeared healthy, but Integument: No ectoparasites were noted. The haircoat was
she had always been thin. There had been no vomiting, full and lustrous.
diarrhea, or changes in thirst or urination. Pesto was obtained Eyes, Ears, Nose, and Throat: In the left eye, mild
as a stray kitten, tested negative for FeLV and FIV, and had conjunctivitis with moderate serous ocular discharge was

Continued
508 SECTION 2  Bacterial Diseases

present. Mild corneal opacity was present in the right eye. splenic, and jejunal nodes. Hepatic and sublumbar nodes
No abnormalities detected on examination of the oral cavity. were enlarged to a lesser degree. The echotexture of the
There was no evidence of nasal discharge or abnormal nasal enlarged nodes was homogeneous. The left adrenal gland
airflow. was moderately enlarged, measuring 0.66 cm in thickness.
Musculoskeletal: Body condition score 4.5/9, ambulatory, The right adrenal gland could not be differentiated from
adequate muscle mass. multiple enlarged nodes. No free fluid was identified.
All Other Systems: No clinically significant abnormalities were Microbiologic Testing: In-clinic FeLV antigen and FIV
detected. A full neurologic examination was not performed. antibody ELISA assay: Negative.
Ophthalmologic Examination: Pesto behaved as if visual Feline coronavirus serum antibody IFA assay: Positive at
and was relatively comfortable with mild blepharospasm. 1:25,600.
The menace response, palpebral reflexes, and pupillary Treatment: Pesto was treated with l-lysine (500 mg PO q12h),
light reflexes were normal. There was no evidence of eyelid for possible FHV-1 infection, and doxycycline (10 mg/kg
abnormalities or episcleral or conjunctival injection. Mild PO q24h, immediately followed by a bolus of water), for
chemosis was present bilaterally. There was no aqueous possible chlamydiosis or mycoplasmosis. The owner could
flare, the lenses were normal, and a fundic examination was not administer the l-lysine, only the doxycycline, and
normal. The dorsal third of the right cornea had superficial reported improvement in Pesto’s activity level and appetite
vessels that extended from the limbus and a 2- to 3-mm several days after starting the doxycycline.
central area where ghost vessels were present, which may Additional Diagnostics and Outcome: Pesto was
have represented a previous dendritic ulcer. A rose Bengal evaluated again 2 weeks later for possible lymph node
stain was negative for ulceration. aspiration or biopsy under sedation. Abdo­minal ultrasound
Laboratory Findings: showed persistent abdominal lymphadenomegaly.
CBC: Cytologic examination of an ultrasound-guided aspirate
HCT 31% (30%-50%) of a mesenteric lymph node revealed mild to moderate
MCV 37.8 fL (42-53 fL) lymphoid reactivity. Plasma cells were increased in number
MCHC 34.2 g/dL (30-33.5 g/dL) and were admixed throughout the specimen with rare
Reticulocytes 29,000 cells/µL (7000-60,000 cells/µL) mast cells and eosinophils. Low numbers of vacuolated
WBC 7030 cells/µL (4500-14,000 cells/µL) macrophages and nondegenerate neutrophils were also
Neutrophils 4570 cells/µL (2000-9000 cells/µL) observed. RT-PCR for feline coronavirus RNA on the lymph
Lymphocytes 2179 cells/µL (1000-7000 cells/µL) node aspirate was negative. IFA serology for antibodies to B.
Monocytes 141 cells/µL (50-600 cells/µL) clarridgeiae was negative, whereas serology for antibodies
Eosinophils 70 cells/µL (150-1100 cells/µL) to B. henselae was positive at 1:64. B. henselae was cultured
Basophils 70 cells/µL (0-50 cells/µL) from the blood. Treatment was changed from doxycycline
Platelets clumped but adequate. A few Dohle bodies were to azithromycin, and again there was mild improvement in
seen within neutrophils. Pesto’s attitude. However, several days later the cat became
Serum Chemistry Profile: lethargic again, and her appetite decreased further.
Sodium 146 mmol/L (151-158 mmol/L) After 3 weeks of treatment with azithromycin, Pesto was
Potassium 4.5 mmol/L (3.6-4.9 mmol/L) reexamined. Because of the cat’s fractious temperament,
Chloride 110 mmol/L (117-126 mmol/L) sedation was required. No abnormalities were detected
Bicarbonate 18 mmol/L (15-21 mmol/L) on physical examination. A CBC showed a hematocrit
Phosphorus 5.1 mg/dL (3.2-6.3 mg/dL) of 23%, MCV of 39.2 fL, MCHC of 32.6 g/dL, reticulocyte
Calcium 9.8 mg/dL (9.0-10.9 mg/dl) count of 23,200 cells/µL, 8220 slightly toxic neutrophils/
BUN 20 mg/dL (18-33 mg/dL) µL, 788 band neutrophils/µL, 1239 lymphocytes/
Creatinine 1.0 mg/dL (1.1-2.2 mg/dL) µL, 788 eosinophils/µL, and 298,000 platelets/µL.
Glucose 119 mg/dL (63-118 mg/dL) A biochemistry panel showed similar abnormalities to those
Total protein 12.0 g/dL (6.6-8.4 g/dL) previously present (mild hyponatremia, hypochloremia,
Albumin 2.4 g/dL (2.2-4.6 g/dL) and hyperglycemia and low creatinine), and the globulin
Globulin 9.6 g/dL (2.8-5.4 g/dL) had increased to 12.4 mg/dL, with a total protein of
ALT 42 U/L (27-101 U/L) 14.4 mg/dL. IFA serology and culture for Bartonella were
AST 25 U/L (17-58 U/L) repeated and showed a titer of 1:1024 to B. clarridgeiae and
ALP 8 U/L (14-71 U/L) 1:4096 to B. henselae; Bartonella spp. culture was negative.
GGT 9 U/L (0-4 U/L) An ultrasound-guided biopsy of an enlarged mesenteric
Cholesterol 134 mg/dL (89-258 mg/dL) lymph node was obtained. Histopathology showed a mixed,
Total bilirubin 0.2 mg/dL (0-0.2 mg/dL). dense inflammatory infiltrate composed of neutrophils,
Serum Protein Electrophoresis: Results were consistent with lymphocytes, and plasma cells. Some of the macrophages
a polyclonal gammopathy. present stained positive using immunohistochemistry for
Imaging Findings: feline coronavirus antigen. Treatment with prednisone
Thoracic Radiographs: No clinically significant abnormalities (1 mg/kg PO q12h for 5 days, then 1 mg/kg PO q24h
were noted. thereafter) was initiated. Four days later, the cat became
Abdominal Ultrasound: Lymph nodes were moderately anorectic and tachypneic and was euthanized. A necropsy
enlarged throughout the abdomen, including the gastric, was not performed.
CHAPTER 52  Bartonellosis 509

Diagnosis: Feline infectious peritonitis and concurrent B. Bartonella bacteremia was an incidental finding. Bartonella
henselae bacteremia bacteremia was detected in the face of doxycycline
Comments: Bartonella infection in this cat was initially treatment, which highlights the fact that antimicrobial drug
suspected because of fever and lymphadenopathy, and treatment does not always clear bacteremia. The cat was
infection with B. henselae was confirmed with Bartonella fractious and the owner could not effectively administer the
culture. Ultimately, an alternate diagnosis (FIP) was made l-lysine twice daily, but assured the veterinarian that Pesto
based on the presence of the combination of marked had received the doxycycline. The owner may have been
hyperglobulinemia, a very high feline coronavirus titer, and exposed to Bartonella during treatment. Treatment with
histopathology of a mesenteric lymph node and IHC for azithromycin was followed by a single negative culture, but
coronavirus antigen. There was also minimal response to antibody titers increased, so the Bartonella infection may
antimicrobial drugs with activity against Bartonella. Thus, the have been relatively recent.

SUGGESTED READINGS 16. Pappalardo BL, Correa MT, York CC, et al. Epidemiologic evalua-
tion of the risk factors associated with exposure and seroreactivity
Breitschwerdt EB. Feline bartonellosis and cat scratch disease. Vet to Bartonella vinsonii in dogs. Am J Vet Res. 1997;58:467-471.
Immunol Immunopathol. 2008;123:167-171. 17. Henn JB, Liu CH, Kasten RW, et al. Seroprevalence of antibodies against
Brunt J, Guptill L, Kordick DL, et al. American Association of Feline Bartonella species and evaluation of risk factors and clinical signs asso-
Practitioners 2006 Panel report on diagnosis, treatment, and preven- ciated with seropositivity in dogs. Am J Vet Res. 2005;66:688-694.
tion of Bartonella spp. infections. J Feline Med Surg. 2006;8:213-226. 18. Mandle T, Einsele H, Schaller M, et al. Infection of human CD34+
progenitor cells with Bartonella henselae results in intraerythro-
REFERENCES cytic presence of B. henselae. Blood. 2005;106:1215-1222.
1. Barton AL. Descripcion de elementos endoglobulares hallados en 19. Chomel BB, Boulouis HJ, Breitschwerdt EB, et al. Ecological fitness
los enfermos de fiebre verrucoca. Cron Med Lima. 1909;26:7-10. and strategies of adaptation of Bartonella species to their hosts and
2. Wear DJ, Margileth AM, Hadfield TL, et al. Cat scratch disease: vectors. Vet Res. 2009;40:29.
a bacterial infection. Science. 1983;221:1403-1405. 20. Pulliainen AT, Dehio C. Persistence of Bartonella spp. stealth
3. Breitschwerdt EB. Feline bartonellosis and cat scratch disease. Vet pathogens: from subclinical infections to vasoproliferative tumor
Immunol Immunopathol. 2008;123:167-171. formation. FEMS Microbiol Rev. 2012;36:563-599.
4. Chomel BB, Kasten RW, Floyd-Hawkins K, et  al. Experimental 21. Kordick DL, Brown TT, Shin K, et al. Clinical and pathologic eval-
transmission of Bartonella henselae by the cat flea. J Clin Micro- uation of chronic Bartonella henselae or Bartonella clarridgeiae
biol. 1996;34:1952-1956. infection in cats. J Clin Microbiol. 1999;37:1536-1547.
5. Boulouis HJ, Chang CC, Henn JB, et  al. Factors associated with 22. Guptill L, Slater LN, Wu CC, et al. Evidence of reproductive failure
the rapid emergence of zoonotic Bartonella infections. Vet Res. and lack of perinatal transmission of Bartonella henselae in experimen-
2005;36:383-410. tally infected cats. Vet Immunol Immunopathol. 1998;65:177-189.
6. Foil L, Andress E, Freeland RL, et al. Experimental infection of domes- 23. Guptill L, Slater L, Wu CC, et al. Experimental infection of young
tic cats with Bartonella henselae by inoculation of Ctenocephalides felis specific pathogen-free cats with Bartonella henselae. J Infect Dis.
(Siphonaptera: Pulicidae) feces. J Med Entomol. 1998;35:625-628. 1997;176:206-216.
7. Reis C, Cote M, Le Rhun D, et al. Vector competence of the tick 24. Mikolajczyk MG, O’Reilly KL. Clinical disease in kittens inocu-
Ixodes ricinus for transmission of Bartonella birtlesii. PLoS Negl lated with a pathogenic strain of Bartonella henselae. Am J Vet Res.
Trop Dis. 2011;5:e1186. 2000;61:375-379.
8. Chomel BB, Boulouis HJ, Breitschwerdt EB. Cat scratch disease 25. Chomel BB, Kasten RW, Williams C, et al. Bartonella endocarditis:
and other zoonotic Bartonella infections. J Am Vet Med Assoc. a pathology shared by animal reservoirs and patients. Ann N Y
2004;224:1270-1279. Acad Sci. 2009;1166:120-126.
9. Chomel BB, Abbott RC, Kasten RW, et  al. Bartonella henselae 26. Chomel BB, Wey AC, Kasten RW, et al. Fatal case of endocarditis
prevalence in domestic cats in California: risk factors and asso- associated with Bartonella henselae type I infection in a domestic
ciation between bacteremia and antibody titers. J Clin Microbiol. cat. J Clin Microbiol. 2003;41:5337-5339.
1995;33:2445-2450. 27. Varanat M, Broadhurst J, Linder KE, et al. Identification of Bar-
10. Chomel BB, Boulouis HJ, Maruyama S, et al. Bartonella spp. in pets tonella henselae in 2 cats with pyogranulomatous myocarditis and
and effect on human health. Emerg Infect Dis. 2006;12:389-394. diaphragmatic myositis. Vet Pathol. 2012;49:608-611.
11. Bai Y, Kosoy MY, Boonmar S, et al. Enrichment culture and molec- 28. Varanat M, Travis A, Lee W, et al. Recurrent osteomyelitis in a cat
ular identification of diverse Bartonella species in stray dogs. Vet due to infection with Bartonella vinsonii subsp. berkhoffii genotype
Microbiol. 2010;146:314-319. II. J Vet Intern Med. 2009;23:1273-1277.
12. Diniz PP, Billeter SA, Otranto D, et al. Molecular documentation of 29. Beerlage C, Varanat M, Linder K, et al. Bartonella vinsonii subsp. berk-
Bartonella infection in dogs in Greece and Italy. J Clin Microbiol. hoffii and Bartonella henselae as potential causes of proliferative vascu-
2009;47:1565-1567. lar diseases in animals. Med Microbiol Immunol. 2012;201:319-326.
13. Brenner EC, Chomel BB, Singhasivanon OU, et al. Bartonella infec- 30. Lappin MR, Kordick DL, Breitschwerdt EB. Bartonella spp anti-
tion in urban and rural dogs from the tropics: Brazil, Colombia, Sri bodies and DNA in aqueous humour of cats. J Feline Med Surg.
Lanka, and Vietnam. Epidemiol Infect. 2012:1-8. 2000;2:61-68.
14. Henn JB, Vanhorn BA, Kasten RW, et al. Antibodies to Bartonella 31. Fontenelle JP, Powell CC, Hill AE, et al. Prevalence of serum anti-
vinsonii subsp. berkhoffii in Moroccan dogs. Am J Trop Med Hyg. bodies against Bartonella species in the serum of cats with or with-
2006;74:222-223. out uveitis. J Feline Med Surg. 2008;10:41-46.
15. Solano-Gallego L, Bradley J, Hegarty B, et al. Bartonella henselae 32. Sykes JE, Westropp JL, Kasten RW, et al. Association between Bar-
IgG antibodies are prevalent in dogs from southeastern USA. Vet tonella species infection and disease in pet cats as determined using
Res. 2004;35:585-595. serology and culture. J Feline Med Surg. 2010;12:631-636.
510 SECTION 2  Bacterial Diseases

33. Dowers KL, Hawley JR, Brewer MM, et al. Association of Barton- 53. Morales SC, Breitschwerdt EB, Washabau RJ, et  al. Detection of
ella species, feline calicivirus, and feline herpesvirus 1 infection with Bartonella henselae DNA in two dogs with pyogranulomatous
gingivostomatitis in cats. J Feline Med Surg. 2010;12:314-321. lymphadenitis. J Am Vet Med Assoc. 2007;230:681-685.
34. Belgard S, Truyen U, Thibault JC, et al. Relevance of feline calici- 54. Kitchell BE, Fan TM, Kordick D, et  al. Peliosis hepatis in a
virus, feline immunodeficiency virus, feline leukemia virus, feline dog infected with Bartonella henselae. J Am Vet Med Assoc.
herpesvirus and Bartonella henselae in cats with chronic gingivo- 2000;216:519-523:517.
stomatitis. Berl Munch Tierarztl Wochenschr. 2010;123:369-376. 55. Saunders GK, Monroe WE. Systemic granulomatous disease and
35. Quimby JM, Elston T, Hawley J, et  al. Evaluation of the asso- sialometaplasia in a dog with Bartonella infection. Vet Pathol.
ciation of Bartonella species, feline herpesvirus 1, feline calicivi- 2006;43:391-392.
rus, feline leukemia virus and feline immunodeficiency virus with 56. Cadenas MB, Maggi RG, Diniz PP, et  al. Identification of bacte-
chronic feline gingivostomatitis. J Feline Med Surg. 2008;10:66-72. ria from clinical samples using Bartonella alpha-Proteobacteria
36. Glaus T, Hofmann-Lehmann R, Greene C, et al. Seroprevalence of growth medium. J Microbiol Methods. 2007;71:147-155.
Bartonella henselae infection and correlation with disease status in 57. Diniz PP, Wood M, Maggi RG, et  al. Co-isolation of Bartonella
cats in Switzerland. J Clin Microbiol. 1997;35:2883-2885. henselae and Bartonella vinsonii subsp. berkhoffii from blood, joint
37. Ueno H, Hohdatsu T, Muramatsu Y, et al. Does coinfection of Bar- and subcutaneous seroma fluids from two naturally infected dogs.
tonella henselae and FIV induce clinical disorders in cats? Micro- Vet Microbiol. 2009;138:368-372.
biol Immunol. 1996;40:617-620. 58. Breitschwerdt EB, Hegarty BC, Maggi R, et  al. Bartonella spe-
38. Lappin MR, Breitschwerdt E, Brewer M, et al. Prevalence of Barton- cies as a potential cause of epistaxis in dogs. J Clin Microbiol.
ella species antibodies and Bartonella species DNA in the blood of 2005;43:2529-2533.
cats with and without fever. J Feline Med Surg. 2009;11:141-148. 59. Cross JR, Rossmeisl JH, Maggi RG, et  al. Bartonella-associated
39. Breitschwerdt EB, Levine JF, Radulovic S, et al. Bartonella henselae meningoradiculoneuritis and dermatitis or panniculitis in 3 dogs.
and Rickettsia seroreactivity in a sick cat population from North J Vet Intern Med. 2008;22:674-678.
Carolina. Int J Appl Res Vet Med. 2005;3:287-302. 60. Yager JA, Best SJ, Maggi RG, et al. Bacillary angiomatosis in an
40. Bayliss DB, Steiner JM, Sucholdolski JS, et  al. Serum feline pan- immunosuppressed dog. Vet Dermatol. 2010;21:420-428.
creatic lipase immunoreactivity concentration and seroprevalences 61. Sykes JE, Kittleson MD, Pesavento PA, et al. Evaluation of the rela-
of antibodies against Toxoplasma gondii and Bartonella species in tionship between causative organisms and clinical characteristics of
client-owned cats. J Feline Med Surg. 2009;11:663-667. infective endocarditis in dogs: 71 cases (1992-2005). J Am Vet Med
41. Pearce LK, Radecki SV, Brewer M, et  al. Prevalence of Barton- Assoc. 2006;228:1723-1734.
ella henselae antibodies in serum of cats with and without clini- 62. Duncan AW, Marr HS, Birkenheuer AJ, et al. Bartonella DNA in
cal signs of central nervous system disease. J Feline Med Surg. the blood and lymph nodes of Golden Retrievers with lymphoma
2006;8:315-320. and in healthy controls. J Vet Intern Med. 2008;22:89-95.
42. Leibovitz K, Pearce L, Brewer M, et al. Bartonella species antibod- 63. Barber RM, Li Q, Diniz PP, et al. Evaluation of brain tissue or cere-
ies and DNA in cerebral spinal fluid of cats with central nervous brospinal fluid with broadly reactive polymerase chain reaction for
system disease. J Feline Med Surg. 2008;10:332-337. Ehrlichia, Anaplasma, spotted fever group Rickettsia, Bartonella,
43. Bettenay SV, Lappin MR, Mueller RS. An immunohistochemical and Borrelia species in canine neurological diseases (109 cases).
and polymerase chain reaction evaluation of feline plasmacytic J Vet Intern Med. 2010;24:372-378.
pododermatitis. Vet Pathol. 2007;44:80-83. 64. Varanat M, Maggi RG, Linder KE, et al. Molecular prevalence of
44. Berryessa NA, Johnson LR, Kasten RW, et al. Microbial culture of Bartonella, Babesia, and hemotropic Mycoplasma sp. in dogs with
blood samples and serologic testing for bartonellosis in cats with splenic disease. J Vet Intern Med. 2011;25:1284-1291.
chronic rhinosinusitis. J Am Vet Med Assoc. 2008;233:1084-1089. 65. Cherry NA, Diniz PP, Maggi RG, et  al. Isolation or molecular
45. Fenimore A, Varanat M, Maggi R, et al. Bartonella spp. DNA in detection of Bartonella henselae and Bartonella vinsonii subsp.
cardiac tissues from dogs in Colorado and Wyoming. J Vet Intern berkhoffii from dogs with idiopathic cavitary effusions. J Vet Intern
Med. 2011;25:613-616. Med. 2009;23:186-189.
46. MacDonald KA, Chomel BB, Kittleson MD, et  al. A prospective 66. Hawkins EC, Johnson LR, Guptill L, et al. Failure to identify an
study of canine infective endocarditis in northern California (1999- association between serologic or molecular evidence of Bartonella
2001): emergence of Bartonella as a prevalent etiologic agent. J Vet infection and idiopathic rhinitis in dogs. J Am Vet Med Assoc.
Intern Med. 2004;18:56-64. 2008;233:597-599.
47. Breitschwerdt EB, Kordick DL, Malarkey DE, et  al. Endocarditis 67. Brunt J, Guptill L, Kordick DL, et  al. American Association of
in a dog due to infection with a novel Bartonella subspecies. J Clin Feline Practitioners 2006 Panel report on diagnosis, treatment,
Microbiol. 1995;33:154-160. and prevention of Bartonella spp. infections. J Feline Med Surg.
48. Ohad DG, Morick D, Avidor B, et  al. Molecular detection 2006;8:213-226.
of Bartonella henselae and Bartonella koehlerae from aortic 68. Cockwill KR, Taylor SM, Philibert HM, et al. Bartonella vinsonii
valves of Boxer dogs with infective endocarditis. Vet Microbiol. subsp. berkhoffii endocarditis in a dog from Saskatchewan. Can
2010;141:182-185. Vet J. 2007;48:839-844.
49. Kelly P, Rolain JM, Maggi R, et al. Bartonella quintana endocardi- 69. Chomel BB, Mac Donald KA, Kasten RW, et al. Aortic valve endo-
tis in dogs. Emerg Infect Dis. 2006;12:1869-1872. carditis in a dog due to Bartonella clarridgeiae. J Clin Microbiol.
50. Henn JB, Gabriel MW, Kasten RW, et al. Infective endocarditis in 2001;39:3548-3554.
a dog and the phylogenetic relationship of the associated “Barton- 70. Davenport AC, Mascarelli PE, Maggi RG, et al. Phylogenetic diver-
ella rochalimae” strain with isolates from dogs, gray foxes, and a sity of bacteria isolated from sick dogs using the BAPGM enrichment
human. J Clin Microbiol. 2009;47:787-790. culture platform [Abstract]. J Vet Intern Med. 2012;26:786-787.
51. Gillespie TN, Washabau RJ, Goldschmidt MH, et al. Detection of 71. Pappalardo BL, Brown T, Gookin JL, et al. Granulomatous disease
Bartonella henselae and Bartonella clarridgeiae DNA in hepatic associated with Bartonella infection in 2 dogs. J Vet Intern Med.
specimens from two dogs with hepatic disease. J Am Vet Med 2000;14:37-42.
Assoc. 2003;222:35:47-51. 72. Gould FK, Denning DW, Elliott TS, et al. Guidelines for the diag-
52. Tabar MD, Maggi RG, Altet L, et  al. Gammopathy in a Span- nosis and antibiotic treatment of endocarditis in adults: a report of
ish dog infected with Bartonella henselae. J Small Anim Pract. the Working Party of the British Society for Antimicrobial Chemo-
2011;52:209-212. therapy. J Antimicrob Chemother. 2012;67:269-289.
CHAPTER 52  Bartonellosis 511

73. Biswas S, Maggi RG, Papich MG, et al. Molecular mechanisms of 78. Kaplan JE, Masur H, Holmes KK. Guidelines for preventing oppor-
Bartonella henselae resistance to azithromycin, pradofloxacin and tunistic infections among HIV-infected persons—2002. Recom-
enrofloxacin. J Antimicrob Chemother. 2010;65:581-582. mendations of the U.S. Public Health Service and the Infectious
74. Yamamoto K, Chomel BB, Kasten RW, et  al. Homologous pro- Diseases Society of America. MMWR Recomm Rep. 2002;51:1-52.
tection but lack of heterologous-protection by various species and 79. Oliveira AM, Maggi RG, Woods CW, et al. Suspected needle stick
types of Bartonella in specific pathogen-free cats. Vet Immunol transmission of Bartonella vinsonii subspecies berkhoffii to a vet-
Immunopathol. 1998;65:191-204. erinarian. J Vet Intern Med. 2010;24:1229-1232.
75. Greene CE, McDermott M, Jameson PH, et al. Bartonella henselae 80. Lin JW, Chen CM, Chang CC. Unknown fever and back pain
infection in cats: evaluation during primary infection, treatment, caused by Bartonella henselae in a veterinarian after a needle punc-
and rechallenge infection. J Clin Microbiol. 1996;34:1682-1685. ture: a case report and literature review. Vector Borne Zoonotic
76. Regnery RL, Rooney JA, Johnson AM, et  al. Experimentally Dis. 2011;11:589-591.
induced Bartonella henselae infections followed by challenge exposure 81. Magalhaes RF, Urso Pitassi LH, Lania BG, et al. Bartonellosis as
and antimicrobial therapy in cats. Am J Vet Res. 1996;57:1714-1719. cause of death after red blood cell unit transfusion. Ultrastruct
77. Bradbury CA, Lappin MR. Evaluation of topical application of 10% Pathol. 2009;33:151-154.
imidacloprid–1% moxidectin to prevent Bartonella henselae trans-
mission from cat fleas. J Am Vet Med Assoc. 2010;236:869-873.
CHAPTER 53

Canine Brucellosis
Autumn P. Davidson and Jane E. Sykes

cause systemic disease (such as discospondylitis) in dogs not


Overview of Brucellosis in Dogs used for reproduction. Dogs with B. canis discospondylitis are
First Described: B. canis was first isolated from dogs by Leland typically medium- to large-breed dogs, with body weights that
Carmichael in 1966 (USA)1 range from 14 to 64 kg; dogs have ranged in age from 1 to 8
Causes: Brucella canis; rarely Brucella suis, Brucella melitensis, years, with a median of 4 years in one study.6-9 Of 22 dogs with
or Brucella abortus B. canis discospondylitis known to the authors, 18 (82%) were
intact male dogs, 3 (14%) were female spayed, and 1 (4%) were
Geographic Distribution: Worldwide, but infections are most
male neutered.6,8-10 Three dogs described in the literature with
often reported from the southwestern United States, Cen-
B. canis ophthalmitis did not fit this signalment; one dog was a
tral and South America, China, and Japan
Pomeranian and none were intact males.11
Major Clinical Signs: No signs or infertility, abortion, non- Transmission of B. canis occurs through direct exposure to
painful scrotal enlargement and dermatitis, testicular body fluids that contain sufficient numbers of bacteria to cause
atrophy, lymphadenopathy, splenomegaly, signs related infection (2 × 106 CFU), most often semen, lochia, aborted
to discospondylitis (spinal pain, pelvic limb paresis, or fetuses/placentas, milk, and/or urine. Transmission is therefore
paralysis), uveitis, chorioretinitis, rarely osteomyelitis and primarily oral, nasal, or conjunctival and secondarily venereal
meningoencephalitis. (i.e., through the mucous membranes). Oronasal and conjuncti-
Major Mode of Transmission: Direct exposure to contami- val exposure follows the ingestion or aerosolization of infectious
nated body fluids (oral, nasal, conjunctival, venereal) materials. The aerosol route is especially important in crowded
kennel conditions. Transplacental transmission and direct cuta-
Differential Diagnoses: Bartonellosis, mycobacterial infec- neous inoculation can also occur. Although fomite-associated
tions, chronic vector-borne diseases associated with transmission can occur under appropriate circumstances,
splenomegaly such as ehrlichiosis and babesiosis, coxiel- B. canis is short-lived outside the host and is readily inactivated
losis, other bacterial and fungal causes of discospondyli- by common disinfectants, such as quaternary ammonium com-
tis, intervertebral disc disease, hemic neoplasia (such as pounds, 1% sodium hypochlorite, 70% ethanol, iodine/alcohol
lymphoma or multiple myeloma). Additional differential solutions, and glutaraldehyde.12
diagnoses for dogs with ophthalmitis include deep myco- Members of the Canidae family are the natural hosts of
ses, rickettsial diseases, leptospirosis, and toxoplasmosis. B. canis. Canine brucellosis occurs most commonly as outbreaks
Human Health Significance: B. canis can be transmitted to in large commercial kennels, and less commonly in privately
humans and most often causes mild, influenza-like illness. owned dogs. Although the geographic distribution of canine
brucellosis is not fully understood, outbreaks of canine brucel-
losis have geographic orientation, with increased incidence seen
Etiology and Epidemiology in the southernmost United States and commonly in Mexico,
Central and South America, the People’s Republic of China,
Canine brucellosis is caused by Brucella canis, a small, gram- and Japan. There are sporadic reports of disease in Canada and
negative, non–spore-forming aerobic coccobacillus. Brucella throughout Europe.7,13-17 Australia and New Zealand appear
abortus, Brucella melitensis, and Brucella suis occasionally to be free of the disease. However, because infection with B.
cause canine infections but are comparatively rare.2,3 B. canis canis is chronic and subclinical, the organism can readily be
infections are zoonotic, but other species of Brucella present a imported into regions where the prevalence of disease is low
greater risk to human health.4,5 through the movement of infected dogs. The increased practice
Brucellosis is the primary contagious infectious venereal dis- and success of canine semen processing for exportation (chill-
ease of concern in canine reproduction. Brucella canis causes ing and cryopreservation) for the purpose of artificial insemi-
reproductive failure in both male and female dogs. Screening for nation now makes canine brucellosis a concern worldwide, as
B. canis infection is an important part of the pre-breeding evalu- direct mucosal contact among dogs is no longer necessary for
ation of any dog and should be included in the initial diagnos- transmission.
tics in any case of canine abortion, orchitis, epididymitis, and
apparent infertility in bitches or dogs. Because the incidence of Clinical Features
canine brucellosis is low in many geographic locations, breeder
compliance with regular screening can wane, which makes con- Signs and Their Pathogenesis
tinued veterinarian vigilance important. Notably, neutered and Brucella organisms attach to and penetrate mucous membranes;
maiden or virgin dogs can also be infected. B. canis can also the severity of disease is proportional to the infectious dose. After

512
CHAPTER 53  Canine Brucellosis 513

FIGURE 53-2  Brucella discospondylitis at the T7-T8 junction with ventral spondylosis
FIGURE 53-1  Midterm abortion from a Brucella canis affected bitch diagnosed by and ­collapse of the intervertebral disc space. (Courtesy Anna Park, DVM, and James Lavely,
AGID. Note the variation in fetal development indicating that death occurred at different DVM, DACVIM [Neurology].)
stages. (Courtesy Patricia Olson, DVM, PhD, DACT.)
and urethra and is shed intermittently in the urine. Anti-sperm
replication in regional lymph nodes, bacteremia occurs within antibodies develop in association with brucellosis-induced
7 to 30 days after exposure with subsequent dissemination to epididymal granulomas and further contribute to infertility.
reticuloendothelial cells, the prostate, uterus, and placenta. Pyospermia develops 3 to 4 months postinfection. Chronic
B. canis has a predilection for steroid-dependent (reproductive) infections in either sex can result in uveitis or endophthalmitis,
tissues. The organism survives facultatively within monocytes lymphadenitis, splenomegaly, or discospondylitis; occasionally,
and macrophages of the reticuloendothelial system. Brucella dermatitis and meningoencephalitis have been reported (Figure
organisms can survive and multiply in reticuloendothelial cells 53-2).3,8,11,20 Owners of dogs with discospondylitis most often
because they inhibit the bactericidal myeloperoxidase-peroxide- describe episodic neck pain or lameness, which can occasion-
halide system through the release of 5′-guanosine and adenine. ally be accompanied by lethargy or decreased activity. Bacte-
Early in infection, polymorphonuclear cells and macrophages remia can persist for years, and subclinically infected dogs can
kill intracellular B. canis relatively ineffectually.18 remain infectious for long intervals. Infection of the bone that
Brucella organisms can be found in the rough endoplasmic surrounded a total hip replacement was described in two dogs
reticulum of placental trophoblastic giant cells in an infected 9 and 12 months after surgery.21
bitch’s gravid uterus. Severe necrotizing placentitis with infarc- Large numbers of organisms are shed in the vulvar discharge
tion of the labyrinth region, coagulation necrosis of the chorionic of bitches 4 to 6 weeks postabortion. The highest concentration
villi, and necrotizing arteritis result in fetal death. The organism of organisms is shed in the semen of infected dogs 2 to 3 months
can be found in the gastric contents of aborted fetuses. Necro- postinfection, with lesser amounts for years. Urine can serve as
tizing vasculitis and granulomatous inflammation results in epi- a contaminated vehicle because of the proximity of the urinary
didymal and subsequent testicular and prostatic pathology.3,19 and genital tracts in the dog, with shedding present for months
Brucellosis in dogs is associated with a high morbidity but to years, and is more prevalent in males. Organisms can also be
low mortality. The clinical systemic signs are often subtle and shed in milk.22-24
include suboptimal athletic performance, lumbar pain, lameness, Spontaneous apparent recovery can occur 1 to 5 years
weight loss, and lethargy. The primary clinical sign of canine postinfection, but true clearance of the organism is difficult
brucellosis in the breeding bitch is pregnancy loss, which can to document. Bitches that have been treated with antimicro-
occur early (day 20) in gestation resulting in fetal resorption, or bial drugs can produce normal litters after multiple abortions,
more commonly (75% of dogs) later in gestation (generally 4 to but can remain infectious to their offspring. Dogs may remain
59 days), which results in abortion (Figure 53-1). Bitches with infertile because of irreparable damage to the spermatogenic
pregnancy loss early in gestation can appear to be infertile (fail apparatus.3,23
to conceive) unless early ultrasonographic pregnancy evaluation
is performed to identify pregnancy. Nongravid bitches can show Physical Examination Findings
no signs or can show regional lymphadenopathy (pharyngeal if For dogs with clinical B. canis infections, fever is rarely pres-
orally acquired, inguinal and pelvic if venereally acquired). ent. Affected male dogs may have an enlarged but nonpain-
The primary acute clinical signs of canine brucellosis in the ful scrotum and secondary scrotal pyoderma, which results
male dog involve the portions of the reproductive tract that from excessive licking. Enlargement of the epididymis is often
participate in the maturation, transport, and storage of sper- present. Some chronically infected male dogs have small, soft
matozoa. Epididymitis is common, with associated orchitis and testicles due to testicular atrophy. Other dogs have general-
scrotal dermatitis, and resultant deterioration of semen qual- ized peripheral lymphadenomegaly, and splenomegaly may be
ity and fertility. Chronically, testicular atrophy and infertility detected on abdominal palpation. The most common finding
can occur. The organism can be found in the prostate gland in dogs with discospondylitis is spinal pain.8,9 Other findings
514 SECTION 2  Bacterial Diseases

TABLE 53-1
Diagnostic Assays Available for Brucella Infection in Dogs
Assay Specimen Type Target Performance
Culture Whole blood, urine, disc aspirates or Brucella spp. Confirms infection but is insensitive. Antibiotic
bone fragments, tissue specimens treatment can lead to false-negative results.
obtained at necropsy (e.g., spleen,
uterus, prostate), aborted fetuses
and placental tissue, seminal fluid
Histopathology Tissue specimens obtained by biopsy Brucella spp. IHC may have low sensitivity depending on the
with organism or at necropsy organisms tissue examined, but supports disease causa-
detection using tion when positive and properly performed and
IHC interpreted.
Serology, rapid Serum Antibodies to Rapid and inexpensive. False positives are com-
slide agglu- Brucella spp. mon as a result of cross-reactivity with other
tination test bacterial species and require confirmation with
(RSAT), and an alternative test. Positive results do not imply
mercaptoetha- active infection. False-negative results with RSAT
nol RSAT can occur in dogs with discospondylitis. The
sensitivity of the RSAT may be higher than that
of the ME-RSAT.
Serology, tube ag- Serum Antibodies to See RSAT. Usually positive by 2 weeks after infection.
glutination test Brucella spp. Titers ≥ 1:200 tend to correlate with bacteremia.
Serology, agar gel Serum Antibodies to See RSAT. Requires special expertise and is time-
immunodiffu- Brucella spp. consuming to perform. May not be positive until
sion 12 weeks after infection. Cytoplasmic antigen as-
says are more specific for Brucella spp. antigens.
Serology, indirect Serum Antibodies to See RSAT. Requires expertise to perform and inter-
immunofluores- Brucella spp. pret. May have lower specificity and sensitivity
cent antibody when compared with ELISA assays or the tube
(IFA) agglutination test.
Serology, ELISA Serum Antibodies to Has the potential to be sensitive and specific, but
Brucella spp. assays vary in performance. A standard, accepted
assay is not currently available.
PCR Whole blood, splenic or lymph node Brucella spp. Confirms active infection and has the potential to
aspirates, disc aspirates, tissue DNA be more sensitive than culture. Sensitivity and
specimens obtained at necropsy, specificity may vary depending on assay design,
vaginal swabs, seminal fluid, urine, specimen type, and quality assurance practices
placental materials, or fetuses in the laboratory. Results should be interpreted
with caution and in light of the clinical picture
and the results of serologic tests and/or culture.

IHC, immunohistochemistry.

include lameness, delayed placing reactions, ataxia, pelvic limb suggest brucellosis, such as dermatitis, uveitis, small testicles,
hyperreflexia, and, uncommonly, pelvic limb paresis or tetra- or lymphadenopathy.8
paresis. Meningoencephalitis may result in behavioral changes,
anisocoria, ataxia, circling, or a head tilt. For unknown rea- Diagnosis
sons, ocular abnormalities are often unilateral and include
blepharospasm, conjunctival hyperemia, and evidence of uve- The diagnosis of canine brucellosis is based on suggestive
itis, such as aqueous flare, miosis, iris hyperpigmentation, loss clinical signs and the results of serology; culture of blood,
of the normal contour of the iris surface, hypopyon, posterior urine, or tissues; histopathology; and/or PCR assays. Because
synechiae, and secondary glaucoma.11 Corneal edema, vitre- no single antemortem test has 100% sensitivity, and serologic
ous opacities, swelling and hyperemia of the optic disc, and assays lack specificity, a combination of diagnostic assays is
evidence of multifocal granulomatous chorioretinitis have also often required to make a diagnosis (Table 53-1). When mul-
been described. Ocular abnormalities and discospondylitis are tiple dogs with infertility problems are present in a household
frequently present in the absence of other systemic signs of ill- or kennel, as many dogs as possible should be tested with
ness. However, some, but not all dogs with discospondylitis serology and blood culture to increase the chance of a reliable
have other, subtle physical examination abnormalities that diagnosis.
CHAPTER 53  Canine Brucellosis 515

A B
FIGURE 53-3  Radiographic images from a 1-year-old female spayed labradoodle with Brucella canis discospondylitis that was evaluated for cervical pain. CSF analysis revealed a CSF
protein concentration of 31 g/dL, with 3 nucleated cells/µL, 89% of which were small mononuclear cells and 11% of which were large mononuclear cells (marginal lymphocytic pleocyto-
sis). Brucella canis was isolated from 5 of 5 blood culture specimens. Urine culture was negative. Of significance, a B. canis rapid slide agglutination test was negative. The dog was treated
successfully with antimicrobial drugs (doxycycline and clavulanic acid–amoxicillin) and lesions resolved radiographically. A, Plain lateral radiograph of the cervical spine. There is endplate
lucency associated with cranial endplate of C3 at the C2-3 articulation. The C2-3 disc space appears narrowed. B, Computed tomography image. Central lysis of the end plates at C2-3 is
present (arrow), compatible with discospondylitis at C2-3.

Laboratory Abnormalities Brucella. Detached sperm heads, proximal and distal cytoplas-
Complete Blood Count, Serum Biochemical Tests, and Urinalysis mic droplets, and acrosomal deformities are the most common
The results of routine laboratory tests are often within reference morphologic abnormalities. Sperm head-to-head agglutination
ranges in dogs with brucellosis. The CBC may show leukocyto- suggests the presence of anti-sperm antibodies. An absence of
sis due to a neutrophilia.11 Degenerate left shifts, monocytosis, sperm in the ejaculate (azoospermia) can also be found.
and/or lymphopenia have been uncommonly reported.9,20 The
chemistry panel is often normal, but hyperglobulinemia and Diagnostic Imaging
hypoalbuminemia may be present. A lack of abnormalities on Plain Radiography and Advanced Imaging
routine laboratory testing or the presence of splenomegaly or Plain radiography may show evidence of either multifocal
abdominal lymphadenomegaly in dogs with discospondylitis or focal discospondylitis (see Figure 53-2; Figure 53-3, A).
should raise suspicion for B. canis infection. Urinalysis is gen- Advanced imaging (CT or MRI) may be more sensitive than
erally within normal limits, even when B. canis bacteriuria is plain radiographs for detection of endplate lucency (see Figure
present. 53-3, B; see also Chapter 85).

Cytologic Analysis of Tissue Aspirates Sonographic Findings


Aspirates of the spleen or lymph nodes of dogs with brucellosis Abdominal ultrasound examination of dogs with brucellosis
reveal lymphoid reactivity, sometimes with plasmacytosis. Pros- may show no abnormalities, or abdominal lymphadenomegaly
tatic aspirates in one dog with prostatitis showed granulomatous and splenomegaly may be present. Ultrasound of the pros-
inflammation with intracellular gram-negative coccobacilli.7 tate may reveal prostatic irregularity, hyperechogenicity, and
cavitation.7
Cytologic Analysis of Body Fluids
CSF analysis in dogs with Brucella discospondylitis is usually Microbiologic Tests
unremarkable. A dog with meningoencephalitis had neutro- Serologic Diagnosis
philic pleocytosis and increased CSF protein concentration.3 There are two reasons to perform serologic tests for brucellosis
Aspirates of ocular fluids (aqueous or vitreous humor) from a in dogs: (1) as part of a routine screening program in breed-
dog with panophthalmitis revealed pyogranulomatous inflam- ing kennels, whereby healthy dogs are screened for infection,
mation with extracellular and intracellular small gram-negative and (2) for diagnosis of active infection in sick dogs that are
cocci.11 suspected to have brucellosis because of infertility, reproduc-
tive losses, epididymitis, discospondylitis, osteomyelitis, or
Semen Examination ophthalmitis.
Diminished sperm counts (oligospermia), poor motility (asthe- Assays used for screening purposes are usually rapid and
nospermia), and increased morphologic abnormalities (tera- inexpensive, but can suffer from low specificity (a high rate of
tospermia) are characteristic of semen in dogs infected with false-positive test results) due to strong cross-reactivity between
516 SECTION 2  Bacterial Diseases

surface lipopolysaccharide (LPS) antigens of B. canis and those other disease manifestations suggestive of brucellosis should be
of other nonpathogenic infectious agents. Up to 50% to 60% of confirmed using culture or a reliable PCR assay whenever pos-
dogs can have false-positive test results because of cross-reacting sible. Although false-negative serologic test results are uncom-
antibodies to microorganisms such as Bordetella, Pseudomonas, mon in dogs with discospondylitis, negative serologic test
Escherichia coli, and Moraxella spp. Even when the antibod- results in dogs with discospondylitis do not rule out infection
ies detected truly represent anti-Brucella antibodies, the pres- with Brucella spp., and attempts should also be made to detect
ence of these antibodies does not indicate active infection with the organism with culture or PCR assays. Of various dogs with
Brucella because dogs can remain seropositive for months after B. canis discospondylitis or ophthalmitis, 15 of 18 were positive
they clear the infection. Antibodies to Brucella do not become with the RSAT, 16 of 16 were positive with the TAT, and 6 of
detectable until 2 to 12 weeks after infection, so a window of 7 were positive with AGID assays.6,8-11 The 3 dogs that were
time exists in which an infected dog eludes serologic diagno- negative with the RSAT had discospondylitis. One was positive
sis with any assay used. False negatives also occasionally occur by blood culture, and the other 2 dogs tested positive with the
in dogs with chronic sequestered infections. Beyond the first 3 TAT at a later date.8,10
months of infection, the sensitivity of screening assays is gener-
ally believed to be high, but in some scenarios it might be lower Isolation and Identification
than previously thought.25 Examples of screening assays include Despite improvements in serologic diagnostic methods, con-
the rapid slide agglutination test (RSAT); the semiquantitative firmatory blood cultures are still indicated when brucellosis is
2-mercaptoethanol modified RSAT (ME-RSAT); the semiquan- suspected. B. canis is readily isolated from the blood of bacte-
titative tube agglutination test (TAT); agar gel immunodiffusion remic individuals for several months after infection. Specimens
(AGID) assays; and indirect IFA or ELISA assays. Veterinar- for blood culture should be collected as detailed in Chapter
ians need to understand the limitations of the assays available 3. A positive B. canis culture has been advocated as the best
to them through local veterinary diagnostic laboratories. The diagnostic test in the first 2 months of the disease; however,
RSAT uses Brucella ovis as the antigen, whereas the ME-RSAT dogs can become abacteremic 27 to 64 months after infection
uses B. canis antigen, which may improve specificity. However, but remain infected. Occasionally B. canis can be isolated from
in one study, the sensitivity of the ME-RSAT was only 32%, urine, tissue specimens such as aborted fetuses or placental
when compared with 71% for the RSAT, so the RSAT may material, necropsy specimens (especially spleen, prostate, and
be preferable for screening.25 In the TAT, a titer of more than uterus), vaginal swabs, or disc aspirates. Of dogs with Brucella
1:200 correlates well with positive blood cultures, but lower discospondylitis or ophthalmitis, 6 of 15 dogs tested positive by
titers are more difficult to interpret. The performance of AGID blood culture, 3 of 13 by urine culture, and 4 of 6 by culture of
assays requires trained laboratory personnel and special media. disc material or bone chips from the intervertebral disc space
Two types of AGID assay are available: an assay that uses cell obtained at surgery.6,8-10
wall antigens as the substrate (cell wall AGID assay) and an B. canis is an aerobe, but unlike other Brucella species, added
assay that uses cytoplasmic antigens of Brucella as the substrate CO2 (5% to 10%) can be inhibitory. Multiplication is slow at
(cytoplasmic AGID assay). The cell wall assay has from the same the optimum temperature of 37°C, and enriched medium (tryp-
low specificity as other assays that detect antibodies to Brucella tose or trypticase soy media) is occasionally needed to support
LPS, and the sensitivity may be lower than that of the RSAT.25 adequate growth. Brucella colonies become visible in 2 to 3 days.
In contrast, the cytoplasmic assay is specific for antibodies to Growth of Brucella can be identified on the basis of rough (as
Brucella, so, when available, it is a useful confirmatory assay for opposed to smooth) colony morphology, staining characteris-
dogs that test positive with other rapid serologic assays. Indi- tics, and slide agglutination with anti-Brucella serum. Identifica-
rect IFA has lower sensitivity than many other serologic assays. tion to the species level is best done in a specialized laboratory.
A number of ELISA assays have been developed to overcome Differentiation between B. canis and B. suis can be difficult.13 In
difficulties that relate to the low specificity of serologic assays the United States, the New York State Diagnostic Laboratory at
for B. canis and the limited availability of other assays. The Cornell University, the Tifton Veterinary Diagnostic and Inves-
performance of these ELISA assays has varied, but sensitive and tigational Laboratory in Georgia, and the University of Florida
specific ELISA assays have been developed that may prove use- are recognized to be reliable for definitive testing made neces-
ful for rapid serologic testing in the future.26 sary by positive B. canis screening tests in dogs intended for
In practice, for screening purposes, healthy bitches are tested breeding, or for clinically affected dogs undergoing evaluation
at least 1 month before a planned breeding or introduction to a for infertility or abortion, or when outbreaks are being man-
breeding kennel with a serologic assay of low specificity such as aged.3,27 Susceptibility testing can be performed, but in vitro test
the RSAT because these assays are rapid, readily available, and results may not correlate well to the in vivo treatment response.
inexpensive. Healthy, fertile stud dogs should be tested annually
with the RSAT. If the test result is negative, in cases of subfertil- Molecular Diagnosis Using the Polymerase Chain Reaction
ity or if recent exposure is suspected, a second test is performed PCR assays have been used to successfully identify B. canis in
a month later, to assess for seroconversion (which would be semen from dogs with negative culture results, which suggests
expected if the dog was early in the course of infection when that this may ultimately be the most sensitive method of testing as
first tested). If the test result is positive, a confirmatory assay part of the male dog pre-breeding evaluation, especially in cases
should be performed. This could include the TAT or a more spe- where management of an outbreak is the concern.28 PCR assays
cific assay, such as the cytoplasmic AGID assay, a specific ELISA were also used to detect B. canis in the aqueous humor of dogs
assay, blood culture, or a reliable PCR assay, although blood with endophthalmitis11 and in lymph node aspirates.29 Assays
culture can lack sensitivity.13,15 Usually the assay(s) selected is have been designed that differentiate between the Brucella species
based on test availability and financial resources of the owner. present.30 A real-time PCR assay was used to detect B. canis in tis-
Positive serologic test results for dogs with discospondylitis or sue specimens, urine, buffy coat, and intervertebral disc material
CHAPTER 53  Canine Brucellosis 517

of a dog with discospondylitis when culture of intervertebral disc treatment spaced 1 month apart, or treatment with streptomy-
material, blood, and urine was negative.7 Some veterinary diag- cin every other week for 8 weeks.11 Combination therapy with
nostic laboratories in the United States now offer real-time PCR tetracyclines and streptomycin is thought to be the most suc-
assays for detection of B. canis on a commercial basis. cessful treatment, but unavailability of streptomycin, nephro-
toxicity, parenteral therapy requirements, and expense may be
Pathologic Findings problematic.31-33 In addition, aminoglycosides do not provide
The main pathologic findings of B. canis infections consist of adequate ocular or CNS penetration for dogs with ophthalmitis
lymphoplasmacytic to lymphohistiocytic follicular hyperplasia or meningitis. A combination of doxycycline and rifampin has
in lymphoid tissues, lymphohistiocytic to neutrophilic endo- been used to treat human brucellosis but may not be well tol-
phthalmitis, and mild lymphohistiocytic meningitis. Occasion- erated by dogs because of gastrointestinal adverse effects. The
ally, mild lymphoid and histiocytic infiltrates are present in other use of three or four drugs in combination (e.g., streptomycin,
tissues, such as the liver and lung.15 A mixed lymphohistiocytic enrofloxacin, doxycycline with or without rifampin) was effi-
inflammatory response is also present in the uterus (endometri- cacious for treatment of ophthalmitis in dogs.11 Topical 1%
tis) of females and the testicles, epididymis and spermatic cord, prednisolone acetate and atropine ointment may be required for
and prostate of male dogs. Prostatic and testicular fibrosis may management of uveitis.11
be found. Placental trophoblasts can contain large numbers of One study reported a possible benefit of therapy with enro-
gram-negative bacteria, and coagulative necrosis may be pres- floxacin (5 mg/kg q12h PO for 4 weeks, often for multiple
ent. In male dogs, lesions are more likely to be restricted to the courses) in a small group of infected dogs and bitches. Enro-
reproductive system. Immunohistochemistry has been used to floxacin did not completely eliminate B. canis, but it maintained
confirm the presence of Brucella spp. in placental tissues.13 fertility and avoided the recurrence of abortions, transmis-
sion of the disease to subsequently whelped puppies, and dis-
Treatment and Prognosis semination of microorganisms during parturition. Ultimately,
however, most treated individuals remained culture positive.34
In certain jurisdictions, brucellosis is a reportable disease Combinations of enrofloxacin and doxycycline may be a more
in dogs or in humans. Infected dogs and bitches should be effective alternative for dogs unable to tolerate aminoglycoside
removed from breeding programs and quarantined. Eradication or rifampin treatment and for dogs that have ocular or CNS
of the disease in kennel situations has not been successful with- involvement, but studies are lacking.
out removal (culling) of all current or historically infected dogs.
Because of the zoonotic potential of the disease and difficulty in Vaccination and Immunity
eradicating the infection, euthanasia of affected dogs has been
recommended. Infection in household or small hobby kennel Serum agglutinating antibodies do not protect against infec-
dogs often results in client requests for alternatives to euthana- tion in the dog, and immunity likely depends on cell-mediated
sia. Neutering decreases the amount of organism shed in semen immunity. Presently, the development of a vaccine is consid-
and uterine discharge, but does not eradicate infection. Urine ered undesirable because the Brucella vaccines evaluated have
shedding can persist, and the organism can be found in internal offered only moderate protection, and immunized dogs develop
organs and the bloodstream. antibodies that confound serodiagnosis.
Antibiotic therapy has historically not been rewarding, likely
because the organism is intracellular and bacteremia is peri- Prevention
odic. Antimicrobial drug treatment can reduce antibody titers
without clearing infection. Relapses are common. Combination Prevention of infection and elimination of infected dogs should
therapy should be used whenever possible, preferably with tet- be the principal control strategy in kennels. Prevention requires
racyclines (high-dose doxycycline or minocycline) for at least 1 at least yearly testing of all breeding stock and the testing of
to 2 months and an aminoglycoside (streptomycin or gentami- all dogs to be introduced into a kennel. Ideally, two negative
cin) for the first 1 to 2 weeks of treatment (Table 53-2). Some screening tests performed at least a month apart should occur
also advocate use of two 1-week courses of aminoglycoside before a dog or bitch is introduced into a breeding facility. Con-
firmed positive dogs should be isolated (euthanasia is advised by
many authors), neutered, treated, and tested monthly (AGID,
TABLE 53-2 culture, or reliable PCR assay) until two consecutive negative
tests occur, although occult infection can still be present.
Medications That Could Be Used to Treat Brucella spp. Infections Private breeders should require serologic screening of all
in Dogs bitches offered for breeding and confirmatory negative test
results if positive results occur during screening before they
Interval accept a bitch into their kennel. Stud dogs should be screened at
Drug Dose (mg/kg) Route (hours) least annually. Because of the potential for nonvenereal trans-
Doxycycline* 10-15 PO 12 mission, screening of maiden dogs and bitches before breeding
Streptomycin 20 IM 24 is also recommended.35,36
Gentamicin sulfate 9-14 IV, IM, SC 24
Rifampin 5 PO 12 Public Health Aspects
Enrofloxacin 5-10 PO 24
Humans can become infected with B. canis, although they
*Use in combination with one of the other drugs listed. Combinations are relatively resistant to infection and other Brucella species
of drugs are preferred for treatment where possible. account for the majority of human disease. Approximately 40
518 SECTION 2  Bacterial Diseases

cases of human infection have been reported worldwide; how- AGID performed at a university veterinary diagnostic labora-
ever, the actual number is unknown. Serologic assays used for tory yielded a negative result, so brucellosis was not thought
diagnosis of brucellosis in humans often do not detect antibod- to be present. However, subsequent blood culture of all of the
ies to B. canis, so the disease may be overlooked. Transmis- dogs in the household was positive in one other dog.37 Affected
sion to humans most commonly occurs through contact with an humans are typically treated successfully with combinations of
infected dog’s semen, vulvar discharge from an infected bitch, or doxycycline and either aminoglycosides (for the first week) or
aborted fetuses or placentas, or through direct, accidental labo- rifampin, but monotherapy has been associated with unaccept-
ratory exposure.8 Thus, disease is more likely to occur in people able rates of relapse.
who are in contact with breeding dogs, but disease occasionally Prevention of human disease involves eradication of the dis-
occurs in the owners of pet dogs with discospondylitis.10 Immu- ease in dog kennels; precautions such as the wearing of gloves
nocompromised humans, children, and those who are pregnant and protective clothing when in contact with aborted mate-
may be at greater risk for development of disease. Infection of rial; and hand washing after handling dogs and before eating.
humans with B. canis typically results in nonspecific, persistent Although it is preferable that immunocompromised individu-
clinical signs of fever, muscle aches, night sweats, and general als do not participate in dog breeding activities, if necessary,
malaise. Rarely, granulomatous hepatitis, hepatosplenomeg- these individuals should take special care if they interact with
aly, osteomyelitis, pulmonary disease, and endocarditis have breeding dogs. Contact with dogs with reproductive problems
been reported. In one case report, disease occurred in an HIV- or their secretions should be avoided whenever possible, and if
infected dachshund dog breeder who reported infertility in one aborted material must be handled, gloves, gowns, and face pro-
of the dogs, which had been tested by a veterinarian and was tection should be worn, all protective wear should be disposed
seropositive with a screening assay. Confirmatory testing with of properly, and hands should be washed thoroughly.

CASE EXAMPLE Brucella canis IFA, which was positive at a titer of 1:200. A
B. canis AGID was submitted and was also positive.
Treatment: Sadie was treated with tramadol (1-2 mg/
Signalment: “Sadie”, a 6-year-old female spayed Border kg PO q8h) and carprofen (3 mg/kg PO q12h). Once
collie mix from northern California the serologic test results were available, a tentative
History: Sadie was brought to her local veterinarian for diagnosis of B. canis discospondylitis was made, and
listlessness and reluctance to jump noted during the she was treated with enrofloxacin (5.5 mg/kg PO q12h).
previous 48 hours. She had been adopted from a local Clinical improvement was reported in 2 weeks, and
humane shelter as an ovariohysterectomized 11-month- repeat radiographs were performed in 1 and 5 months.
old. She lived in a rural agricultural area in northern Equivocal change was reported in the appearance of T7-
California and was the only dog in the household. T8 at 1 month, but the 5-month radiograph showed no
Physical Examination: Sadie’s vital signs were within progression, suggesting the lesion was no longer active.
normal limits, mucous membrane color was pink, The owners were advised of the zoonotic potential of this
and she was quiet and alert on physical examination. disease and the potential hazard of transmission to other
Body weight was 25 kg. Mild dental tartar was evident. dogs. The potential need for lifelong antibiotic therapy
Thoracic auscultation and abdominal palpation were with risk of relapse was also discussed.
normal, pulses were synchronous, and no peripheral Comments: Dogs with Brucella discospondylitis frequently
lymphadenomegaly was evident. Palpable dorsal are brought to their veterinarian for clinical signs of spinal
thoracolumbar pain was demonstrated. She was reluctant pain or lameness that resemble common musculoskeletal
to rise but was ambulatory. The neurologic examination injuries such as intervertebral disc disease or rupture of
including a fundoscopic examination was normal. the cranial cruciate ligament. Although not typical, this
Laboratory Findings: A complete blood count, chemistry case illustrates the fact that B. canis disease can occur in
panel, and urinalysis were performed. Mild neutropenia female spayed, nonbreeding dogs and often occurs in the
(2958 cells/µL; reference range, 3000-11,500 cells/µL) was absence of fever or significant abnormalities on routine
found, and the chemistries and urinalysis were normal laboratory testing. A diagnosis of Brucella discospondylitis
with no proteinuria. was strongly suspected based on positive serologic
Imaging Findings: Radiographs of the spine revealed test results with multiple assays. Blood culture was not
evidence of disc space collapse, spondylosis, and performed, but urine culture was negative. Monotherapy
discospondylitis at the T7-T8 disc space (see Figure 53-2). with fluoroquinolones has been associated with high
Microbiologic Testing: A SNAP 4DX test (IDEXX rates of relapse in human patients once antimicrobial
Laboratories) for antigen of Dirofilaria immitis and drug treatment has been discontinued, so it is generally
antibodies to Ehrlichia canis, Anaplasma phagocytophilum, not recommended. Nevertheless, it can occasionally be
and Borrelia burgdorferi was negative. Aerobic bacterial successful.
urine culture was negative. Serum was submitted for a
CHAPTER 53  Canine Brucellosis 519

SUGGESTED READINGS 19. Moore JA, Kakuk TJ. Male dogs naturally infected with Brucella
canis. J Am Vet Med Assoc. 1969;155:1352-1358.
Hollett RB. Update on canine brucellosis. Theriogenology. 2010;1: 20. Dawkins BG, Machotka SV, Suchmann D, et al. Pyogranulomatous
287-295. dermatitis associated with Brucella canis infection in a dog. J Am
Lawaczeck E, Toporek J, Cwikla J, et  al. Brucella canis in a HIV- Vet Med Assoc. 1982;181:1432-1433.
infected patient. Zoonoses Public Health. 2011;58:150-152. 21. Smeak DD, Olmstead ML, Hohn RB. Brucella canis osteomyelitis
Wanke MM. Canine brucellosis. Anim Reprod Sci. 2004;82-83:195- in two dogs with total hip replacements. J Am Vet Med Assoc.
207. 1987;191:986-990.
22. Carmichael LE, Kenney RM. Canine abortion caused by Brucella
REFERENCES canis. J Am Vet Med Assoc. 1968;152:605-616.
1. Carmichael LE. Abortion in 200 beagles. J Am Vet Med Assoc. 23. Johnson CA, Walker RD. Clinical signs and diagnosis of Brucella
1966;149:1126. canis infection. Compend Cont Educ Pract Vet. 1992;14:763-772.
2. Barr SC, Eilts BE, Roy AF, et al. Brucella suis biotype 1 infection in 24. Schoeb TR, Morton R. Scrotal and testicular changes in canine bru-
a dog. J Am Vet Med Assoc. 1986;189:686-687. cellosis: a case report. J Am Vet Med Assoc. 1978;172:598-600.
3. Greene CE, Carmichael LE. Canine brucellosis. In: Greene CE, ed. 25. Keid LB, Soares RM, Vasconcellos SA, et al. Comparison of agar
Infectious Diseases of the Dog and Cat. 4th ed. St. Louis, MO: gel immunodiffusion test, rapid slide agglutination test, microbio-
Elsevier Saunders; 2012:398-411. logical culture, and PCR for the diagnosis of canine brucellosis. Res
4. Blankenship RM, Sanford JP. Brucella canis. A cause of undulant Vet Sci. 2009;86:22-26.
fever. Am J Med. 1975;59:424-426. 26. de Oliveira MZ, Vale V, Keid L, et  al. Validation of an ELISA
5. Lucero NE, Escobar GI, Ayala SM, et al. Diagnosis of human brucel- method for the serological diagnosis of canine brucellosis due to
losis caused by Brucella canis. J Med Microbiol. 2005;54:457-461. Brucella canis. Res Vet Sci. 2011;90:425-431.
6. Anderson GI, Binnington AG. Discospondylitis and orchitis associ- 27. Wooley RE, Hitchcock PL, Blue JL, et  al. Isolation of Brucella
ated with high Brucella titre in a dog. Can Vet J. 1983;24:249-252. canis from a dog seronegative for brucellosis. J Am Vet Med Assoc.
7. Corrente M, Franchini D, Decaro N, et  al. Detection of Brucella 1978;173:387-388.
canis in a dog in Italy. New Microbiol. 2010;33:337-341. 28. Keid LB, Soares RM, Vasconcellos SA, et al. A polymerase chain
8. Kerwin SC, Lewis DD, Hribernik TN, et al. Diskospondylitis asso- reaction for the detection of Brucella canis in semen of naturally
ciated with Brucella canis infection in dogs: 14 cases (1980-1991). infected dogs. Theriogenology. 2007;67:1203-1210.
J Am Vet Med Assoc. 1992;201:1253-1257. 29. Aras Z, Ucan US. Detection of Brucella canis from inguinal
9. Henderson RA, Hoerlein BF, Kramer TT, et  al. Discospondylitis lymph nodes of naturally infected dogs by PCR. Theriogenology.
in three dogs infected with Brucella canis. J Am Vet Med Assoc. 2010;74:658-662.
1974;165:451-455. 30. Kang SI, Her M, Kim JW, et  al. Advanced multiplex PCR assay
10. Sykes JE. Unpublished observations, 2012. for differentiation of Brucella species. Appl Environ Microbiol.
11. Ledbetter EC, Landry MP, Stokol T, et  al. Brucella canis endo- 2011;77:6726-6728.
phthalmitis in 3 dogs: clinical features, diagnosis, and treatment. 31. Jennings PB, Crumrine MH, Lewis Jr GE, et al. The effect of a two-
Vet Ophthalmol. 2009;12:183-191. stage antibiotic regimen on dogs infected with Brucella canis. J Am
12. Carmichael LE, Joubert JC. Transmission of Brucella canis by con- Vet Med Assoc. 1974;164:513-514.
tact exposure. Cornell Vet. 1988;78:63-73. 32. Johnson CA, Bennett M, Jensen RK, et al. Effect of combined anti-
13. Brennan SJ, Ngeleka M, Philibert HM, et al. Canine brucellosis in biotic therapy on fertility in brood bitches infected with Brucella
a Saskatchewan kennel. Can Vet J. 2008;49:703-708. canis. J Am Vet Med Assoc. 1982;180:1330-1333.
14. Dunne J, Sehgal K, McMillan A, et al. Canine brucellosis in a dog 33. Nicoletti P. Further studies on the use of antibiotics in canine bru-
imported into the UK. Vet Rec. 2002;151:247. cellosis. Compend Cont Educ Pract Vet. 1991;13:944.
15. Gyuranecz M, Szeredi L, Ronai Z, et  al. Detection of Brucella 34. Wanke MM, Delpino MV, Baldi PC. Use of enrofloxacin in the
canis–induced reproductive diseases in a kennel. J Vet Diagn Invest. treatment of canine brucellosis in a dog kennel (clinical trial). The-
2011;23:143-147. riogenology. 2006;66:1573-1578.
16. Nockler K, Kutzer P, Reif S, et  al. Canine brucellosis—a case 35. Hollett RB. Canine brucellosis: outbreaks and compliance. Therio-
report. Berl Munch Tierarztl Wochenschr. 2003;116:368-372. genology. 2006;66:575-587.
17. Strom Holst B, Lofqvist K, Ernholm L, et al. The first case of Bru- 36. Jones RL, Emerson JK. Canine brucellosis in a commercial breed-
cella canis in Sweden: background, case report and recommen- ing kennel. J Am Vet Med Assoc. 1984;184:834-835.
dations from a Northern European perspective. Acta Vet Scand. 37. Lawaczeck E, Toporek J, Cwikla J, et al. Brucella canis in a HIV-
2012;54:18. infected patient. Zoonoses Public Health. 2011;58:150-152.
18. Shin SJ, Carmichael LE. Canine brucellosis caused by Brucella
canis. In: Carmichael LE, ed. Recent Advances in Canine Infectious
Diseases. Ithaca, NY: International Veterinary Information Service;
1999.
CHAPTER 54

Tetanus and Botulism


Jane E. Sykes

been hypothesized to reflect an adaptive response to a carnivo-


Overview of Tetanus and Botulism rous lifestyle. C. botulinum produces seven related toxins: A, B,
First Described: The etiology of tetanus was identified in the late C, D, E, F, and G. Most human botulism is caused by types A,
1800s in Germany (Nicolaier).1 C. botulinum and its toxin were B, and E.6 Botulism in dogs and cats almost always results from
first described in Belgium in 1897 (van Ermengem).2 Before ingestion of toxin C,3,5,7-9 with the exception of a few dogs in
that, descriptions of both diseases had been recorded; teta- the Senegal that had type D intoxication.10
nus was described by Hippocrates more than 2000 years ago.
Causes: Toxins of Clostridium botulinum (botulism) or Clos-
Tetanus
tridium tetani (tetanus) C. tetani is widespread in the soil and feces of animals. It pro-
duces two toxins, tetanospasmin and tetanolysin. Tetanolysin
Geographic Distribution: Worldwide
does not appear to have clinical significance. Humans and horses
Major Mode of Transmission: Ingestion of preformed toxin are most susceptible to tetanus, and so widespread vaccination
(botulism) or cutaneous inoculation of spores into a against the disease is performed in these species. Disease occurs
wound (tetanus) occasionally in dogs and is rare in cats. The development of
Major Clinical Signs: Spastic paralysis (tetanus) or flaccid tetanus in dogs and cats usually follows introduction of spores
paralysis (botulism) into a penetrating wound, but more than one third of affected
dogs have no known wound history (cryptogenic tetanus).11
Differential Diagnoses: Differential diagnoses for suspected Traumatic wounds of the feet, claws, or head; draining tracts
botulism include polyradiculoneuritis, paralytic rabies, tick associated with grass awn migration; animal bite wounds; tick
paralysis, coral snake poisoning, myasthenia gravis, and bite wounds; bleeding claw trim wounds; wounds that result
ionophore toxicity. For tetanus they include strychnine toxi- from chronic protozoal or fungal infections of the skin; teething
cosis, hypocalcemia, metaldehyde toxicity, and extraocular wounds in dogs; and surgical wounds (which include spay and
myositis neuter operations) have all been implicated.11-14 Although dogs
Human Health Significance: Botulism and tetanus are not of any age, breed, or sex develop tetanus, disease occurs most
directly transmissible from animals to humans, but care often in young, large-breed, active, intact male dogs.11,13,14 Dis-
should be taken when handling specimens from animals ease is seen in puppies as young as 8 weeks of age; some studies
with botulism. describe a significant proportion of affected dogs as less than
1 year of age. Disease is also more severe in young dogs.11 Cats
are often young cats with outdoor access, but cats of any age
have been affected.15
Etiology and Epidemiology
Clinical Features
Tetanus and botulism are neurologic diseases caused by the
toxins of Clostridium tetani and Clostridium botulinum that Signs and Their Pathogenesis
occur worldwide. These organisms are gram-positive, sluggishly The toxins of C. tetani and C. botulinum have similar structures
motile, anaerobic spore-forming bacilli. The spores of C. tetani and mechanisms of action, even though they cause diseases that
and C. botulinum are extremely resistant in the environment contrast sharply with one another. Both toxins are among the
and resist boiling and disinfection with alcohol or formalin. most potent toxins known and act at femtomolar concentra-
tions. They consist of two polypeptide chains, a heavy (H) chain
Botulism and a light (L) chain, joined by a disulfide bond (Figure 54-1).
In dogs and especially cats, botulism is rare and usually follows The heavy chain attaches to presynaptic nerve terminal mem-
ingestion of preformed neurotoxin within carrion, especially brane receptors, although the precise receptors have not been
waterfowl or poultry carrion. Disease in dogs can coincide with identified because of the extremely low concentrations at which
outbreaks of botulism in waterfowl. Botulism outbreaks in dogs these toxins act. Variation in receptor binding affinities may
have been reported in hunting foxhounds.3,4 Most affected dogs explain differences in species susceptibility to clostridial neu-
are medium- to large-breed, intact male or intact female dogs rotoxins. The toxin is then internalized via receptor-mediated
that vary in age. The only report of botulism in cats was an out- endocytosis into the presynaptic nerve terminal, where it has a
break of disease that occurred after several cats were fed pelican local impact on nerve cell function (botulinum toxin) and also
carrion.5 The relative resistance of dogs and cats to botulism has travels by retrograde axonal transport to the nerve cell body

520
CHAPTER 54  Tetanus and Botulism 521

lower motor neurons and is internalized, travels up axons by


retrograde axonal flow, and enters inhibitory interneurons in
the brain and spinal cord, where it interferes with release of
γ-aminobutyric acid (GABA) and glycine inhibitory neurotrans-
mitters (Figure 54-2, B). Toxin can also spread hematogenously
to the central nervous system (CNS) from the wound site. The
result is unhindered excitation of motor neurons, spastic paraly-
sis, and dysfunction of the sympathetic and parasympathomi-
metic nervous systems. Clinical signs occur 3 days to 3 weeks
after a wound in both cats or dogs. Shorter incubation periods
occur when the wound is proximal to the CNS and when a large
amount of toxin is present. Paralysis may initially be localized
FIGURE 54-1  Schematic of the structure of Clostridium tetani and Clostridium botu- to the wound site but can then generalize. Involvement of the
linum toxins. head is common in dogs, and often owners of dogs with tetanus
first notice an unusual appearance to their dog’s eyes, a “swol-
and diffuses into inhibitory interneurons, where it impacts len” face, or evidence of dysphagia.11 Regurgitation or vomit-
inhibitory interneuron function (tetanospasmin). The L chain, ing of food occurs in less than 20% of affected dogs, and a
a zinc-dependent matrix metalloproteinase, is released from the voice change or dysuria may be noted. Death occurs in severely
vesicle and subsequently cleaves one or more docking proteins. affected dogs due to paralysis of respiratory muscles; infectious
These proteins are critical for the release of neurotransmitters complications such as pneumonia; upper airway obstruction;
from synaptic vesicles into the synaptic cleft and are located the development of severe hyperthermia with disseminated
on the membrane of synaptic vesicles (synaptobrevin) or the intravascular coagulation (DIC), multiple organ failure, and/or
presynaptic cell membrane (syntaxin and SNAP-25). Synapto- pulmonary thromboembolism; and cardiac arrhythmias.11,13,14
brevin binds to the docking proteins located on the presynaptic Hiatal hernia and coxofemoral joint luxations have also been
membrane. The resulting complex (the SNARE complex) docks described as complications of tetanus in dogs.11,14,17 Disease in
the synaptic vesicle at the proper location for fusion and exo- cats is usually localized to the limbs, but generalized tetanus
cytosis of neurotransmitter molecules into the synaptic space can occur.15,18-21 Many affected cats have an initial history of
(Figure 54-2). Tetanospasmin cleaves synaptobrevin, whereas disappearance for days to a week or more, after which lameness
botulinum neurotoxin C cleaves syntaxin; other botulinum is noted.
neurotoxins cleave synaptobrevin or SNAP-25. The process of
recovery involves the sprouting of new presynaptic terminals, Physical and Neurologic Examination Findings
followed by recovery of the original synapse, after which new Dogs and cats with botulism or tetanus are usually alert, afe-
terminal outgrowths retract.16 brile, and in good body condition, but lethargy and anorexia
occur in some animals. Hyperthermia can occur with severe
Botulism tetanus.
C. botulinum spores germinate in anaerobic conditions and pro-
duce toxin. They grow best in temperatures that range from Botulism
30°C to 37°C. The toxin is ingested, survives the acid stomach Physical examination findings in dogs with botulism depend
conditions, and is absorbed into the bloodstream from the small on the severity of intoxication. Fever is absent unless aspi-
intestine. It then travels to peripheral cholinergic synapses, ration pneumonia has developed. Mildly affected dogs show
which include the neuromuscular junction and autonomic syn- only ataxia or pelvic limb paresis, whereas dogs with severe
apses, and inhibits release of acetylcholine (ACh). This leads to intoxication are quadriplegic with absent voluntary motor
ascending symmetrical flaccid paralysis, development of mega- activity and are unable to rise from lateral recumbency. The
esophagus, and signs of autonomic dysfunction. Clinical signs ability to wag the tail is often retained.3,8,9 Neurologic exami-
usually occur within 12 to 72 hours after ingestion of botulinum nation can reveal decreased cranial nerve function, especially
toxin and may be complicated by signs of dietary indiscretion, facial nerve function and a decreased gag reflex, sometimes
although gastrointestinal signs may also reflect the presence of accompanied by ptyalism.3,7,9,22 A weak blink reflex can result
autonomic dysfunction. Disease progresses rapidly over 1 or 2 in corneal ulceration. A mucopurulent ocular discharge may
days, and then begins to resolve with supportive care. Death can be present. Mydriasis with sluggish pupillary light reflexes
result from respiratory muscle paralysis or aspiration of intesti- has been described.3,22 Segmental reflexes may be intact,
nal contents. Abnormalities in affected cats were ascending flac- decreased, or absent, but pain sensation is preserved. Occa-
cid paralysis, lethargy, hypothermia, tachypnea, dehydration, sionally tachycardia and tachypnea are present. Tachypnea
and urinary incontinence.5 may result from diaphragmatic muscle weakness or second-
ary aspiration pneumonia. Increased lung sounds may also
Tetanus be present on thoracic auscultation of dogs with secondary
C. tetani spores are introduced into tissues where they germinate, aspiration pneumonia. Physical examination findings in one
replicate locally, and produce toxin in anaerobic conditions. affected cat consisted of hypothermia, lethargy, dehydration,
Ingestion is not a route of transmission, because tetanospasmin quadriplegia, and decreased segmental reflexes.5
is destroyed by gastrointestinal secretions. The toxin is produced
as a single polyprotein that is cleaved extracellularly by a bacte- Tetanus
rial protease into the L and H chains, which remain connected In dogs with tetanus, involvement of the head and generalized
by the disulfide bond. It then binds to presynaptic terminals of disease occurs more often than occurs in cats. A wound may or
522 SECTION 2  Bacterial Diseases

Skeletal Muscle

Normal Acetylcholine Exocytosis Disruption of Acetylcholine Exocytosis


at the Neuromuscular Junction at the Neuromuscular Junction

Ingestion
of toxin
Botulinum
toxin Light chain

s s

Heavy chain Absorption of


toxin from
intestine
Light chain of
botulinum toxin
cleaves syntaxin
and
interferes with release
of acetylcholine

Acetylcholine
Neurotransmitter Toxin into
vesicle Vesicle and Vesicle and blood vessel
SNARE
proteins terminal terminal
Synaptic fusion membranes membranes
VAMP/ complex forms fuse fail to fuse
Synaptobrevin
SNAP-25
Syntaxin

Synaptic Cleft

Heavy chain of
botulinum toxin
binds to receptor
on cell membrane

1A Acetylcholine Receptors Stimulated 1B Acetylcholine Receptors Not Stimulated


A (Normal Muscle Contraction) (Muscle Fails to Contract - Flaccid Paralysis)
FIGURE 54-2  Action of clostridial neurotoxins. A, Botulism Toxin.
Continued

may not be found. Dogs with localized forelimb tetanus have mouth. Dogs that are ambulatory walk slowly with a stiff, stilted
caudal retraction of the limb with extension of both the elbow gait and can have a wide-based or “sawhorse” stance; voluntary
and carpus.15 In dogs with generalized tetanus, a wrinkled fore- movement of the head may occur slowly or appear difficult, and
head, erect ears, retracted lips (risus sardonicus), and prolapsed the tail may be erect. Laryngeal spasm and hypersalivation occur
third eyelids are almost always present, usually in association in approximately half of affected dogs.13,14 Reflexes are exag-
with generalized muscle stiffness (see Figure 54-3, A). Miosis gerated or difficult to elicit. Severely affected dogs are anxious
may be present. It may be difficult or impossible to open the and hypersensitive, develop opisthotonos, vocalize, tremble or
CHAPTER 54  Tetanus and Botulism 523

Inhibitory
interneuron

Clostridium tetani

internalized
neurotoxin

Toxin into
blood vessel

TeNT

Synaptic Cleft

B
FIGURE 54-2, cont’d.  B, Tetanospasmin.

even have a seizure when stimulated, and may be hyperthermic episodes of dyspnea. At least a third of dogs are bradycardic or
due to increased muscle activity (up to 111°F or 43.9°C).11 A have bradyarrhythmias (which include atrioventricular block or
semicomatose state may even result. Abdominal palpation may sinus arrest), with heart rates that range from 40 to 60 beats/
reveal an enlarged bladder that is difficult to express, or palpa- min; another third of dogs are tachycardic (>150 beats/min) or
tion may not be possible because of abdominal muscle spasm. have variable heart rates, which may reflect autonomic dysfunc-
Hypoventilation may be present, or tetanic spasms may trigger tion or pain.11
524 SECTION 2  Bacterial Diseases

Cats with localized tetanus often have extensor rigidity of may be deviated toward the affected limb.15 When disease gen-
a single limb, on which a wound may be identified. When the eralizes, the contralateral limb and subsequently all four limbs
forelimb is affected, spasm of the triceps muscle results in caudal may be affected.20 Affected cats may be reluctant to move or
extension of the limb with carpal flexion (in contrast to dogs) hyperesthetic. Lockjaw (trismus), protrusion of the third eyelid,
and elbow extension (Figure 54-3, B).20,23,24 The head and neck retracted lips (risus sardonicus), wrinkling of the forehead, or
opisthotonos can occur in cats with generalized tetanus. Seg-
mental reflexes may be exaggerated, or it may not be possible
to elicit reflexes because of severe muscle spasticity. Dyspnea,
tachypnea, and open-mouth breathing may occur.12 Cranial
nerve responses are usually normal. Miosis may be present.

Diagnosis
Diagnosis of tetanus and botulism in dogs and cats is most often
based on a consistent history and clinical abnormalities, exclusion
of the few other possible causes of disease (e.g., with assays for
myasthenia gravis in dogs with botulism), and sometimes electro-
diagnostic testing. The primary differential diagnoses for botulism
are acute polyradiculoneuritis and myasthenia gravis; paralytic
rabies should also be considered. There are few other disorders
other than strychnine poisoning that truly mimic tetanus. Serologic
assays have been used to detect an antibody response to botulinum
A toxins. Finally, specific assays that detect botulinum toxin in serum,
gastrointestinal contents, or food can be performed (Table 54-1).

Laboratory Abnormalities
Complete Blood Count, Serum Biochemical Tests, and Urinalysis
Routine laboratory test results in animals with tetanus or botulism
are often normal, but stress hyperglycemia, mild to severe increases
in the activity of serum creatine kinase (usually <20,000 U/L but
sometimes >200,000 U/L, reference range 46-320 U/L), mildly
increased AST activity, and myoglobinuria may be present in animals
with tetanus.11,14 Dehydration may result in hemoconcentration or
prerenal azotemia. Animals with wounds or aspiration pneumonia
may have an inflammatory leukogram. Severe hyperthermia in dogs
with tetanus may lead to laboratory evidence of multiple organ dys-
function, with moderate to severe hypoalbuminemia, azotemia,
B and/or increased liver enzyme activities and hyperbilirubinemia.
FIGURE 54-3  A, Image of a Weimaraner with generalized tetanus. The dog walked Thrombocytopenia and coagulation test abnormalities consistent
with a stiff gait and was salivating profusely. Risus sardonicus is evident. B, Three-year-old with DIC may also be present in these dogs.
female domestic shorthair cat with localized tetanus that was seen for a 3-day history of
right thoracic limb lameness. Extension of the shoulder and elbow and partial flexion of CSF Analysis
the carpal joint is present. (From Langner KFA, Schenk HC, Leithaeuser C, et al. Localised CSF analysis in dogs and cats with botulism and tetanus
tetanus in a cat. Vet Rec 2011;169:126.) ­typically shows no abnormalities.

TABLE 54-1
Laboratory Assays Available for Diagnosis of Botulism
Assay Specimen Type Target Performance
Mouse inoculation Feces, vomitus, intesti- Botulinum toxin Gold standard for diagnosis. False negatives can occur
­bioassay nal contents, serum, if toxin degradation occurs. Slow turnaround time
foodstuffs means diagnosis is generally retrospective. Laborious
and requires the use of laboratory animals.
ELISA toxin assays Feces, vomitus, intesti- Botulinum toxin Rapid and inexpensive. Standard ELISA toxin assays
nal contents, serum, have a lower sensitivity than the mouse inoculation
foodstuffs assay.
ELISA assays that detect Serum Anti–botulinum Negative antibody titers occur early in the course of
the antibody response toxin antibodies disease, so acute and convalescent titers must be
to botulinum toxin performed. Thus, diagnosis is retrospective.
CHAPTER 54  Tetanus and Botulism 525

pneumonia, although EMG can be performed without anesthe-


sia.3 Electroneurography studies involve electrical stimulation of
one or more motor nerves and assessment of the resultant electri-
cal activity in the innervated muscle. Electroneurography findings
in dogs with botulism include normal F waves (late waves that
disappear in acute polyradiculoneuritis) and decreased magnitude
of the compound muscle action potential.3,22,25 Repetitive nerve
stimulation at low frequencies can result in decremental responses,
but repetitive nerve stimulation at high frequencies in dogs with
botulism has no effect on the muscle response.22 In humans with
botulism, an incremental response to repetitive nerve stimulation
occurs at high frequencies.26 Motor nerve conduction velocity
(MNCV) is normal or decreased. On EMG, the primary find-
ing is prolonged or increased insertional activity in botulism.3,22
Fibrillation potentials and positive sharp waves have also been
described.3,25 Animals with tetanus have had normal EMGs and
MNCVs as well as motor unit potentials and pathologic spon-
A
taneous muscle activity,14,20,21,24 but electrodiagnostic testing is
not specific for a diagnosis of tetanus and is usually not required.

Microbiologic Tests
Isolation and Identification
Attempts to culture C. tetani from wounds are not recommended
because the presence of C. tetani in a wound does not necessar-
ily imply toxin production. In addition, this is insensitive.11,13

Serologic Diagnosis
ELISA assays that detect the antibody response to botulinum neu-
rotoxin are available and were used to confirm a diagnosis of bot-
ulism in one dog.7 A convalescent specimen collected 3 weeks after
the onset of illness was positive, but specimens collected at the time
the dog was first evaluated and 10 days later were negative. Thus,
the diagnosis is generally retrospective with this method because
most dogs have recovered by the time seroconversion occurs. The
B antibody response to tetanospasmin is not significant.
FIGURE 54-4  Radiographic findings in dogs with botulism (A) and tetanus (B). A, Lat-
eral thoracic radiograph from a 6-year-old female spayed Labrador retriever with botulism. Toxin Detection
Megaesophagus is present. The white arrows delineate the dorsal and ventral esophageal Botulinum toxin can be detected in feces, vomitus, gastrointesti-
wall. B, Lateral thoracic radiograph from a 3-year-old female spayed English springer span- nal contents collected at necropsy, carrion, or serum. Specimens
iel with tetanus that was seen for vomiting, dysuria, respiratory distress, and a stiff gait. should be held at 4°C, marked with a warning label, and shipped
Pneumothorax and a hiatal hernia (arrows) can be appreciated. Before the radiographs immediately to the laboratory. The most sensitive assay is a mouse
were taken, an exploratory laparotomy had been performed because a foreign body was inoculation assay, in which untreated mice and mice treated with
suspected. The pneumothorax developed postoperatively for reasons that were unclear. A serotype-specific antitoxin are inoculated with the specimen to
foxtail wound was later found on the left distal pelvic limb between the digits. The dog was determine if the antitoxin is protective.27 However, several days
hospitalized for 19 days, during which time all radiographic abnormalities resolved.
are required to run this assay. Toxin type–specific ELISA assays
are available for rapid detection of neurotoxin C, but standard
Diagnostic Imaging ELISA assays lack sensitivity when compared with mouse inocu-
Thoracic radiographs in dogs with botulism may be unremark- lation. Research efforts have been focused on the development of
able, or megaesophagus may be present (Figure 54-4, A). Evi- a variety of novel, rapid, highly sensitive, and inexpensive diag-
dence of carrion bones are occasionally present in the stomach. nostic assays that detect minute quantities of botulinum toxins.27
Changes consistent with aspiration pneumonia in dogs with
botulism or tetanus may also be apparent.11,22 Rarely, dogs with Pathologic Findings
tetanus have evidence of a hiatal hernia on thoracic radiogra- There are no specific gross or histopathologic findings associ-
phy, which appears as a soft tissue mass in the caudal thorax ated with botulism or tetanus. Evidence of complications such
(see Figure 54-4, B). Abdominal ultrasound examination may be as aspiration pneumonia, pulmonary thromboembolism, or
normal, or mild abdominal lymphadenopathy may be identified. DIC may be present in dogs with tetanus.

Electrodiagnostic Tests Treatment and Prognosis


Electromyography (EMG) and electroneurography can be useful
to support a diagnosis of botulism in some dogs (Table 54-2). Specific Treatment
Sedation or anesthesia for electrophysiologic testing is gener- There is no readily available specific treatment for botulism
ally not advisable in dogs with megaesophagus and aspiration in dogs. In the United States, heptavalent equine antitoxin is
526 SECTION 2  Bacterial Diseases

TABLE 54-2
Pathophysiologic, Clinical, and Electrophysiologic Features of Botulism, Polyradiculoneuritis, and Myasthenia Gravis
Botulism Polyradiculoneuritis Acquired Myasthenia Gravis
Pathophysiology Inhibition of acetylcholine release by Inflammatory disorder of Antibody formation to acetylcho-
botulinum toxin nerve roots line receptors at the neuromus-
cular junction
Neurologic Decreased reflexes, progressive Decreased reflexes, progres- Normal reflexes, muscle weak-
­abnormalities ­tetraparesis, respiratory compro- sive tetraparesis, facial ness and rapid fatigue,
mise, autonomic signs, dysphonia, weakness, dysphonia, respi- ­megaesophagus
­megaesophagus ratory compromise, with or
without hyperesthesia
EMG Normal/subnormal Abnormal Normal
Electroneurography Decreased CMAP amplitudes, normal Decreased CMAP amplitudes, Usually normal CMAP ampli-
F-waves, decremental response to absent F-waves, usually tudes, decremental response to
repetitive nerve stimulation at low normal MNCV repetitive nerve stimulation at
frequency (and incremental response low frequency
at high frequency in humans)

Modified from Uriarte A, Thibaud JL, Blot S. Botulism in 2 urban dogs. Can Vet J 2010;51:1139-1142.
CMAP, Compound muscle action potential; MNCV, Motor nerve conduction velocity.

TABLE 54-3 The efficacy of tetanus antitoxin is controversial, and the opti-
mal dose is not known. In one study, early IV administration of
Suggested Dosages of Medication for Specific Treatment equine tetanus antitoxin did not influence disease progression
of Tetanus or mortality as opposed to administration later in the course of
disease.11
Interval Duration For animals with tetanus, treatment with antimicrobials that
Drug Dose Route (hours) (days) have activity against C. tetani is also recommended. This is not
Metronidazole 15 mg/kg IV, PO 12 10 indicated for animals with botulism, because ingested toxin is
preformed. Appropriate antimicrobials for treatment of tetanus
Tetanus equine 100-1000 IV, IM* NA Once
include intravenous metronidazole or penicillin G. Metronida-
antitoxin units/kg NA Once
zole is preferred because of its improved penetration into puru-
500 units SC near
lent material and the fact that penicillin can antagonize GABA.
wound site
Studies in human patients have also shown improved outcome
Human tetanus 500 units IM near IM Once with metronidazole as opposed to procaine penicillin.29 Once
immune wound if antitoxin has been administered and spasms are controlled (see
globulin found Supportive Care), wounds should also be debrided or flushed
extensively with hydrogen peroxide solution. Earlier appropri-
NA, Not applicable.
ate antimicrobial treatment and wound debridement may not
*IV use is preferred. Administer 0.1 mL intradermally as a test dose be-
fore infusion. The full dose should then be given slowly over 10 minutes. influence progression or mortality.11

Supportive Care
available from the Centers for Disease Control and Prevention For both tetanus and botulism, supportive care is an essential
for treatment of human botulism. However, its availability is part of management. The extent of care required depends on
tightly controlled, so it may be impossible to obtain for treat- the severity of illness. Intravenous fluids, soft bedding, frequent
ment of dogs or cats.28 Tetanus antitoxin is more widely avail- turning to prevent decubital ulcers, application of ocular lubri-
able and should be administered as a single dose IV (equine cant to prevent corneal ulceration, bladder expression or uri-
antitoxin) or IM (human antitoxin) after an initial intradermal nary catheterization (ideally intermittent), enemas to relieve
test dose to evaluate for allergic reactions (Table 54-3). Dogs constipation, and nutritional support in the form of hand feed-
that react to the test dose should be premedicated with diphen- ing, feeding tubes, or total parenteral nutrition may be required.
hydramine and a low dose of dexamethasone. Adverse reactions Force feeding is not recommended because it is unlikely to
to antitoxin include vomiting, retching, or tachypnea and may satisfy caloric needs and can contribute to the development of
occur even in premedicated dogs.11 Reduction in the rate of aspiration pneumonia. Occasionally (<10% of affected dogs
antitoxin administration or discontinuation of treatment leads with tetanus), mechanical ventilation is required to support res-
to resolution of clinical signs. Administration of antitoxin does piration.11 For dogs with botulism, antimicrobial drugs should
not reverse existing paralysis because it is unable to enter nerve only be used if infection develops (e.g., bacterial pneumonia or
terminals, but it may neutralize free toxin. A small amount of urinary tract infection). Aminoglycosides should be avoided
antitoxin can be administered locally if a wound is identified. because they may contribute to neuromuscular blockade.
CHAPTER 54  Tetanus and Botulism 527

Dogs or cats with generalized tetanus should be placed in


a quiet, dimly lit environment away from excessive visual or TABLE 54-4
auditory stimuli. Cotton balls can be used as earplugs. Seda- Tetanus Severity Classification System for Dogs
tives and/or muscle relaxants such as acepromazine, diazepam,
midazolam, or methocarbamol may be useful. Severely affected Class Clinical Signs
animals may require a constant-rate infusion of drugs such as
pentobarbital, phenobarbital, or propofol. Barbiturates are no I Absence of class II, III or IV signs and any or all of:
longer recommended for treatment of tetanus in humans; ben- • Miosis, enophthalmos, risus sardonicus, erect
zodiazepines are preferred. If human disease is severe, drugs that ears, or trismus
cause neuromuscular blockade such as vecuronium are used, but • Hypersensitivity to noise, light or touch
this necessitates mechanical ventilation.30 Methocarbamol often • Ambulatory
appears ineffective and requires frequent administration.11,13 II Absence of class III or IV signs and any or all of:
Benzodiazepines are preferred to methocarbamol because they • Dysphagia
are GABA agonists and also provide some sedation. Success- • Stiff gait, sawhorse stance, erect tail
ful control of muscle spasms relieves pain and hyperthermia. • Ambulatory
If used, opioids should be administered with caution because III Class I or class II signs, absence of class IV signs, and
they may contribute to constipation or gastrointestinal adverse any or all of:
effects. The use of fans and cool fluids may also be beneficial • Recumbency
for dogs with tetanus-induced heat stroke. Administration of • Muscle fasciculations or spasms
atropine may be required for animals with bradyarrhythmias • Seizures
(HR <60 beats/min). Rarely, upper respiratory obstruction has
IV Class I, II, or III signs and any or all of:
necessitated tracheostomy,14 or bradyarrhythmias have required
• Bradycardia (HR ≤60 beats/min) or bradyar-
temporary pacemaker placement.11
rhythmia
Prognosis • Sinus tachycardia (HR ≥140 beats/min) or
tachyarrhythmia
Provided complications do not occur, prognosis is generally
• Labile hypertension or hypotension (mean
good for animals with tetanus or botulism. The overall 28-day
arterial blood pressure ≤60 mm Hg or ≥ 130
survival rate for dogs with tetanus in one study was around
mmHg, or systolic arterial blood pressure ≤80
80%,11 whereas in another study of 20 dogs it was only 50%.13
mm Hg or ≥150 mm Hg)
A classification system has been developed to assess the sever-
• Periods of apnea or respiratory arrest
ity of tetanus in dogs for the purpose of prognostication (Table
54-4).11 The survival rate for dogs that progressed to tetanus Modified from Burkitt JM, Sturges BK, Jandrey KE, et al. Risk factors
severity classes I or II was 100%, but it dropped to 60% for associated with outcome in dogs with tetanus: 38 cases (1987-2005).
dogs that progressed to tetanus severity class III or IV.11 How- J Am Vet Med Assoc 2007;230:76-83.
ever, at the time of initial evaluation, only 11% of 37 dogs were
in classes III or IV, and dogs progressed to higher classes unpre-
dictably, with 47% of the dogs categorized in class III or IV to pets. The spores of C. botulinum are inactivated by pressure-
when the highest recorded class was assigned. Dogs with severe cooking foods.
tetanus are more likely to develop complications such as aspira- For tetanus, control of (1) roaming, (2) exposure to plant
tion pneumonia or multiple organ dysfunction than dogs with awns, and (3) aggressive interactions with other animals may
mild tetanus. In a few dogs that recover (<10% of dogs that reduce the chance that wounds develop. If wounds are noticed,
develop tetanus), intermittent localized muscle spasm continues they should be cleaned promptly with soap and water. Surgical
to occur for many months, which may be noticed by the owner instruments should always be sterilized properly before use so
during activities such as sleeping or eating.11 that tetanus spores are inactivated, and needles should never be
reused. Tetanus spores can be inactivated with iodine, glutaral-
Immunity and Vaccination dehyde, hydrogen peroxide, or autoclaving at 121°C and 15 psi
for 15 minutes.
Given the relative rarity of tetanus and botulism in dogs and
cats, vaccination is not recommended for these species. Recur- Public Health Aspects
rent tetanus or botulism has been documented in human
patients, so recovery from natural disease does not necessarily Animals with botulism or tetanus cannot transmit these condi-
result in protection from future episodes of disease. Thus, pre- tions directly to humans who handle them. Nevertheless, feces,
ventative measures should be discussed with owners of pets that tissues, or body fluids from animals with botulism should be
recover from tetanus or botulism. handled with care and marked with warning labels before they
are submitted to the laboratory because the toxin is so potent.
Prevention The main risk that dogs with tetanus pose to humans is through
bite wounds, which can occur when owners attempt to admin-
Prevention of botulism involves limiting access of cats and dogs ister medications.
to carrion and feeding heat-processed commercial diets that are Three forms of botulism occur in people: infant botulism,
unspoiled. Although botulinum toxin can be denatured through wound botulism, and food botulism.26 Infant botulism occurs as a
boiling, spores can survive this treatment. Thus, cooked food result of local production of botulinum toxin within the intestinal
should not be allowed to stand for long periods before it is fed tract by C. botulinum in the absence of the competing intestinal
528 SECTION 2  Bacterial Diseases

microflora that are normally found in adults. Wound botulism Significant concerns have been raised about the possible use of
occurs almost exclusively as a result of injection of contaminated botulinum toxin as an agent of bioterrorism. This has stimulated
heroin into the skin by intravenous drug users. Botulinum toxin considerable research on the disease, which is otherwise very rare.
preparations are used to treat conditions such as strabismus, Tetanus in humans is rare in developed countries, because
esophageal achalasia, cervical dystonia, axillary hyperhydrosis most children are immunized against the disease. When it
(excessive sweating), or blepharospasm. There are rare reports occurs, it is most often in people older than 60 years of age
of iatrogenic botulism after such use.31 Life-threatening iatro- because of waning immunity.30 Worldwide, the vast majority
genic botulism has also occurred after use of botulinum prepara- of cases occur in neonates from sub-Saharan Africa because of
tions that were unlicensed for cosmetic treatment of wrinkles.32 lack of immunization.

CASE EXAMPLE Segmental Reflexes: Reduced muscle tone was present in the
thoracic limbs. Triceps, patella, and gastrocnemius reflexes
were decreased. Reduced thoracic limb withdrawal reflexes
Signalment: “Jenny,” a 6-year-old female spayed Labrador were noted, but pelvic limb withdrawal reflexes were intact.
retriever from Winters in northern California Panniculus and perineal reflexes were also intact. Pain
History: Jenny was brought to the University of California, perception remained intact.
Davis, veterinary emergency service early in the morning for a Spinal Palpation: No apparent pain was present on palpation
2-hour history of inability to stand. Her owners awoke to find or cervical manipulation.
her in another room, unable to get up for her morning walk. Neuroanatomic Location: Neuromuscular disease.
She appeared anxious and was breathing with increased Laboratory Findings:
effort, but was still wagging her tail. She also did not want CBC:
to eat breakfast. There had been no known history of travel, HCT 41.0% (40%-55%)
toxins, trauma, or tick exposure. Over the past 2 months, MCV 69 fL (65-75 fL)
she had been treated with prednisone (1 mg/kg PO q24h MCHC 36.6 g/dL (33-36 g/dL)
for 5 days, followed by 0.74 mg/kg PO q24h thereafter) for
WBC 5800 cells/µL (6000-13,000 cells/µL)
allergic dermatitis. Three weeks before the onset of illness she
Neutrophils 3671 cells/µL (3000-10,500 cells/µL)
was treated with a short course of cephalexin after an aural
Lymphocytes 1769 cells/µL (1000-4000 cells/µL)
hematoma was repaired. Jenny had been regularly vaccinated
Monocytes 197 cells/µL (150-1200 cells/µL)
with rabies, distemper, hepatitis, and parvovirus vaccines.
Eosinophils 145 cells/µL (0-1500 cells/µL)
Physical Examination:
Platelets clumped but adequate (150,000-400,000 platelets/µL).
Body Weight: 27.2 kg
General: Bright, alert, responsive, and hydrated. Laterally Serum Chemistry Profile:
recumbent and anxious. HR = 140 beats/min, T = 101°F, Sodium 146 mmol/L (145-154 mmol/L)
panting. Potassium 3.9 mmol/L (3.6-5.3 mmol/L)
Integument, Eyes, Ears, Nose, and Throat: Sutures were Chloride 107 mmol/L (108-118 mmol/L)
present on the pinna of the right ear from the recent aural Bicarbonate 21 mmol/L (16-26 mmol/L)
hematoma repair. Moderate dental calculus and gingivitis Phosphorus 2.3 mg/dL (3.0-6.2 mg/dL)
was present. There were no other abnormalities noted. Calcium 11.4 mg/dL (9.7-11.5 mg/dl)
Musculoskeletal: BCS 5/9. The dog was unable to use the BUN 15 mg/dL (5-21 mg/dL)
pelvic limbs to bear weight. Creatinine 0.9 mg/dL (0.3-1.2 mg/dL)
Cardiovascular, Respiratory, Gastrointestinal, Genitouri- Glucose 105 mg/dL (64-123 mg/dL)
nary, and Lymph Nodes: No clinically significant abnor- Total protein 7.1 g/dL (5.4-7.6 g/dL)
malities were present. Albumin 4.2 g/dL (3.0-4.4 g/dL)
Neurologic Examination: Globulin 2.9 g/dL (1.8-3.9 g/dL)
Mentation: Appropriate, anxious. ALT 64 U/l (19-67 U/L)
Gait/Posture: The dog was nonambulatory, but voluntary AST 25 U/L (19-42 U/L)
movement was present in all four limbs. She was unable to ALP 129 U/L (21-170 U/L)
stand and support weight, but could crawl and shuffle. She Creatine kinase 57 U/L (51-399 U/L)
attempted to bear weight on her thoracic limbs and could GGT 4 U/L (0-6 U/L)
maintain sternal recumbency. When supported to stand, the Cholesterol 204 mg/dL (135-361 mg/dL)
pelvic limbs collapsed and the thoracic limbs were minimally Total bilirubin 0.2 mg/dL (0-0.2 mg/dL).
able to bear weight. Urinalysis: SGr 1.025; pH 7.0, trace protein (SSA), 1+ bilirubin,
Cranial Nerves: Slightly reduced palpebral reflexes and gag no hemoprotein, glucose, ketones, WBC/HPF, RBC/HPF,
reflex. Absent corneal reflexes. No other deficits were noted. bacteria or crystals in the sediment. Many lipid droplets
Postural Reactions: These were difficult to assess because of were present.
the dog’s inability to stand, but appeared decreased.
CHAPTER 54  Tetanus and Botulism 529

Imaging Findings: parenteral nutrition were offered to the owner, but were
Thoracic Radiographs: The cardiopulmonary structures were declined. Four days after admission, improvement in the
within normal limits. There were mineral opacities within the triceps reflex was noted, and on day 5, some voluntary
stomach. There was a small amount of gas in the midthoracic movement was noted in the pelvic and thoracic limbs. At
esophagus. that time Jenny was successfully fed meatballs of canned
Abdominal Ultrasound Examination: Mild mesenteric food by hand. She was kept in sternal recumbency for 30
lymphadenopathy was identified. No other abnormalities minutes after eating to minimize further aspiration. On
were present. day 7, the acetylcholine receptor antibody and botulinum
Neuromuscular Testing: Edrophonium response test for toxin assay results became available.
myasthenia gravis: negative Serum acetylcholine receptor antibody titer: 0.05 nmol/L (refer-
Treatment: Jenny was admitted to the intensive care ence range, <0.6 nmol/L)
unit and treated with supportive care that included IV Botulinum toxin C mouse bioassay (fecal specimen): Positive
fluids (lactated Ringer’s solution with 20 mEq/L KCl at for type C botulinum toxin. An extract from the fecal speci-
75 mL/hr), soft bedding, intermittent manual bladder men was lethal in the bioassay. The effects were eliminated
expression, physical therapy (massage and passive range- by heating the extract at 80°C for 10 minutes, which indi-
of-motion exercises), and frequent turning. Treatment cated that the toxic effects were due to a heat-labile toxin.
with prednisone was discontinued. Respiratory function Mice were also protected from the effects by anti–type
was monitored continuously by observation, serial C botulinum antisera but not by anti–type A botulinum
examination, and serial venous pCO2 measurements. antisera.
After repeated inspections for ticks, fipronil was applied Diagnosis: Botulism.
in case the signs resulted from tick paralysis. Serum was Outcome: On day 8 after hospitalization, thoracic radiographs
submitted for an acetylcholine receptor antibody titer, showed resolution of megaesophagus and no evidence of
and feces were submitted for a botulinum toxin assay. pneumonia. By day 10, Jenny began to shuffle along the floor.
Over the subsequent 12 to 18 hours, Jenny became All segmental reflexes had returned but remained reduced.
anxious after she was fed canned food and began to The dog was discharged from the hospital with instructions
retch and hypersalivate. Thoracic radiographs were for continuation of care in the home environment. By day 15,
repeated and showed megaesophagus (see Figure 54-4, Jenny’s owners reported that her normal bark had returned,
A). Severe tachycardia also developed (HR = 210 beats/ and she could walk 20 steps and defecate and urinate
min). An ECG showed ventricular tachycardia followed unassisted. A month after discharge she was reexamined
by supraventricular tachycardia. Systolic blood pressure and no abnormalities were detected.
was 145 mm Hg. Acepromazine was administered (0.01 Comments: The main differential diagnoses considered for
mg/kg IV once), after which her heart rate decreased this dog were myasthenia gravis and botulism. Initially,
to 90 beats/min, with a normal sinus rhythm. A second polyradiculoneuritis was also considered, but the subsequent
neurologic examination showed progression of disease development of megaesophagus was not consistent
with reduced cervical tone and an inability to hold up the with this disorder. Ultimately, a diagnosis of botulism was
head, tetraplegia with absence of all segmental reflexes, confirmed through assay of the stool for botulinum toxin. A
and a reduced tail wag. The owners were warned that history of carrion ingestion was not known, but the dog lived
mechanical ventilation may be needed should the disease in a semirural area, and the presence of mineralized stomach
progress. Over the course of the next day, dysphonia and contents suggested that it had occurred. Prednisone
a mild cough developed, and Jenny regurgitated several administration may have contributed to polyphagia and
times. Gastrostomy or jejunostomy tube feeding or ingestion of carrion by this dog.

SUGGESTED READINGS
3. Barsanti JA, Walser M, Hatheway CL, et al. Type C botulism in
Burkitt JM, Sturges BK, Jandrey KE, et al. Risk factors associated with American Foxhounds. J Am Vet Med Assoc. 1978;172:809-813.
outcome in dogs with tetanus: 38 cases (1987-2005). J Am Vet Med 4. Darke PG, Roberts TA, Smart JL, et al. Suspected botulism in fox-
Assoc. 2007;230:76-83. hounds. Vet Rec. 1976;99:98-99.
Cai S, Singh BR, Sharma S. Botulism diagnostics: from clinical symp- 5. Elad D, Yas-Natan E, Aroch I, et  al. Natural Clostridium botu-
toms to in vitro assays. Crit Rev Microbiol. 2007;33:109-125. linum type C toxicosis in a group of cats. J Clin Microbiol.
Chertow DS, Tan ET, Maslanka SE, et al. Botulism in 4 adults following 2004;42:5406-5408.
cosmetic injections with an unlicensed, highly concentrated botuli- 6. Sobel J, Tucker N, Sulka A, et al. Foodborne botulism in the United
num preparation. JAMA. 2006;296:2476-2479. States, 1990-2000. Emerg Infect Dis. 2004;10:1606-1611.
7. Bruchim Y, Steinman A, Markovitz M, et al. Toxicological, bacte-
REFERENCES riological and serological diagnosis of botulism in a dog. Vet Rec.
1. Nicolaier A. Über infectiösen Tetanus. Dtsch Med Wochenschr. 2006;158:768-769.
1884;10:842-844. 8. Wallace V, McDowell DM. Botulism in a dog—first confirmed case
2. Van Ermengem EP. Über einen neuen anaeroben Bacillus und in New Zealand. N Z Vet J. 1986;34:149-150.
seine Beziehung zum Botulismus. Z Hyg Infektionskrankh. 9. Farrow BR, Murrell WG, Revington ML, et al. Type C botulism in
1897;26:1-56. young dogs. Aust Vet J. 1983;60:374-377.
530 SECTION 2  Bacterial Diseases

10. Doutre MP. 2nd case of botulism type D in the dog in Senegal. Rev 23. Malik R, Simpson DJ, Church DB. What is your diagnosis? Teta-
Elev Med Vet Pays Trop. 1983;36:131-132. nus. J Small Anim Pract. 1998;39(217):252.
11. Burkitt JM, Sturges BK, Jandrey KE, et al. Risk factors associated 24. De Risio L, Gelati A. Tetanus in the cat—an unusual presentation.
with outcome in dogs with tetanus: 38 cases (1987-2005). J Am Vet J Feline Med Surg. 2003;5:237-240.
Med Assoc. 2007;230:76-83. 25. van Nes JJ, van der Most van Spijk D. Electrophysiological evi-
12. Sykes JE. Unpublished observations, 2012. dence of peripheral nerve dysfunction in six dogs with botulism
13. Bandt C, Rozanski EA, Steinberg T, et  al. Retrospective study type C. Res Vet Sci. 1986;40:372-376.
of tetanus in 20 dogs: 1988-2004. J Am Anim Hosp Assoc. 26. Reddy P, Bleck TP. Clostridium botulinum (botulism). In:
2007;43:143-148. Mandell GL, Bennett JE, Dolin R, eds. Principles and Prac-
14. Adamantos S, Boag A. Thirteen cases of tetanus in dogs. Vet Rec. tice of Infectious Diseases. 7th ed. Philadelphia, PA: Elsevier;
2007;161:298-302. 2010:3097-3102.
15. Malik R, Church DB, Maddison JE, et al. Three cases of local teta- 27. Cai S, Singh BR, Sharma S. Botulism diagnostics: from clinical
nus. J Small Anim Pract. 1989;30:469-473. symptoms to in vitro assays. Crit Rev Microbiol. 2007;33:109-125.
16. Meunier FA, Lisk G, Sesardic D, et al. Dynamics of motor nerve 28. Investigational heptavalent botulinum antitoxin (HBAT) to
terminal remodeling unveiled using SNARE-cleaving botulinum replace licensed botulinum antitoxin AB and investigational
toxins: the extent and duration are dictated by the sites of SNAP- botulinum antitoxin E. MMWR Morb Mortal Wkly Rep.
25 truncation. Mol Cell Neurosci. 2003;22:454-466. 2010;59:299.
17. Goldhammer MA, Chapman PS, Grierson JM. Coxofemoral luxa- 29. Ahmadsyah I, Salim A. Treatment of tetanus: an open study to
tion in a Border collie as a complication of a Clostridium tetani compare the efficacy of procaine penicillin and metronidazole. Br
infection. J Small Anim Pract. 2008;49:159-162. Med J (Clin Res Ed). 1985;291:648-650.
18. Langner KF, Schenk HC, Leithaeuser C, et al. Localised tetanus in 30. Reddy P, Bleck TP. Clostridium tetani (tetanus). In: Mandell GL,
a cat. Vet Rec. 2011;169:126. Bennett JE, Dolin R, eds. Principles and Practice of Infectious Diseases.
19. Lee EA, Jones BR. Localised tetanus in two cats after ovariohyster- 7th ed. Philadelphia, PA: Elsevier; 2010:3091-3096.
ectomy. N Z Vet J. 1996;44:105-108. 31. Coban A, Matur Z, Hanagasi HA, et  al. Iatrogenic botulism
20. Polizopoulou ZS, Kazakos G, Georgiadis G, et al. Presumed local- after botulinum toxin type A injections. Clin Neuropharmacol.
ized tetanus in two cats. J Feline Med Surg. 2002;4:209-212. 2010;33:158-160.
21. Tomek A, Kathmann I, Faissler D, et al. [Tetanus in cats: 3 case 32. Chertow DS, Tan ET, Maslanka SE, et al. Botulism in 4 adults fol-
descriptions]. Schweiz Arch Tierheilkd. 2004;146:295-302. lowing cosmetic injections with an unlicensed, highly concentrated
22. Uriarte A, Thibaud JL, Blot S. Botulism in 2 urban dogs. Can Vet J. botulinum preparation. JAMA. 2006;296:2476-2479.
2010;51:1139-1142.
CHAPTER 55

Yersinia pestis (Plague) and Other Yersinioses


Jane E. Sykes and Bruno B. Chomel

of the intestinal tract, which causes the “starved” flea to repeat-


Overview of Plague edly and aggressively feed and regurgitate bacteria into the bite
First Described: Descriptions of plague date back to around site of a new host.2 A large number of flea species can transmit
1300 bc. The causative agent was isolated in 1894, in Hong Y. pestis, but some flea species, such as the Oriental rat flea
Kong (Alexandre Yersin).1 (Xenopsylla cheopis) worldwide and the ground squirrel flea
Cause: Yersinia pestis, a gram-negative coccobacillus (family (Oropsylla montana) in the western United States are partic-
Enterobacteriaceae) ularly efficient vectors. Although nowhere near as efficient as
Affected Hosts: Y. pestis causes disease in rodents, rabbits, these two fleas as a vector, the cat flea (Ctenocephalides felis)
wild carnivores, domestic dogs and cats, and humans. can infest rodent reservoirs and also has the potential to trans-
Goats, deer, antelope, camelids, and nonhuman primates mit Y. pestis.3 Y. pestis attains very high concentrations in
can also develop illness. rodent blood (108 organisms/mL) before death of the rodent
occurs, which allows fleas to acquire sufficient numbers of
Geographic Distribution: Southwestern United States; foci bacteria for efficient transmission to other rodents.4 Fleas can
also exist in Asia, Africa, and South America remain infected for more than a year, which permits transmis-
Major Mode of Transmission: Flea-borne, via an enormous sion long after the death of an infected rodent.
variety of rodent flea species; ingestion of infected Plague is transmitted to cats or dogs when they ingest infected
rodents or rabbits by cats and dogs mammals (especially rodents or rabbits) or less commonly, after
the bite of a rodent flea. Humans are infected by (1) the bite of an
Major Clinical Signs: Fever, lethargy, inappetence, lymphade-
infected flea; (2) direct contact with contaminated tissues (such
nopathy, subcutaneous abscesses
as handling dead rodents, rabbits, or other dead wild mamma-
Differential Diagnoses: Tularemia, cat bite abscesses, Strepto- lian carnivores); or rarely (3) inhalation of respiratory secretions
coccus spp. infections, feline infectious peritonitis by infected animals, especially cats (Figure 55-2). Death of a
Human Health Significance: A small percentage (10%) of rodent from plague can cause the rodent’s fleas to bite humans
human plague cases result from exposure to infected cats, and other animals in a search for an alternative blood meal.
through bites, scratches, or respiratory droplet inhalation In the United States, plague in humans, dogs, and cats is
from cats with pneumonic plague. Dogs and cats can also now uncommon, although it is likely underrecognized in dogs
expose humans through the carriage of infected fleas into and cats. Cats are considerably more susceptible to disease than
households (especially sleeping areas). dogs. Affected cats can be of any age, breed, and sex, with a
mean age of 3 years in one case series.5 Most plague cases in
humans and cats occur in the southwestern United States, par-
ticularly northern New Mexico, but also southern Colorado,
Etiology and Epidemiology northern Arizona, and to a lesser extent California, southern
Oregon, and western Nevada (especially in the Sierra Mountain
The genus Yersinia includes 11 different species, three of which ranges) (Figure 55-3). The probability of an area in the United
are pathogenic in humans, cats, and dogs: Yersinia pestis and States being a high-risk plague habitat increases with elevation
the enteropathogenic species Yersinia pseudotuberculosis and up to 2300 meters and declines as elevation increases there-
Yersinia enterocolitica. Yersinia spp. are nonmotile, gram-neg- after.6 Rodent or rabbit species associated with human cases
ative coccobacilli that belong to the family Enterobacteriaceae. include white-tailed antelope squirrels, ground squirrels, rock
Only Y. pestis is transmitted by arthropod vectors. squirrels, cottontail rabbits, jack rabbits, prairie dogs, deer mice,
Yersinia pestis causes plague, which is maintained in wild Colorado chipmunks, and wood rats.2 The prevalence of anti-
burrowing rodents in rural regions of Asia, Africa, and the bodies to Y. pestis in a survey of 4115 dogs and 466 cats from
Americas (Figure 55-1). There is no plague in Australia, and the California in the mid-1990s was less than 5% in both species.7
last case of plague in Europe was reported after World War II. In general, plague has a seasonal distribution that correlates
The organism is transmitted between rodents by rodent fleas; with the timing of epizootics that lead to a die-off of susceptible
humans, dogs, and cats are usually accidental and “dead-end” rodents. In the United States, most plague cases occur from Feb-
hosts (although direct transmission can occur through aerosols ruary through August, when rodents and their fleas are most
in humans and cats with pneumonic disease). Fleas become active and humans and their companion animals are more likely
infected when they feed on heavily bacteremic rodents. The to be outdoors. In general, the organism does not survive well in
organism replicates in the flea intestine and creates a blockage the environment outside infected hosts, with survival less than

531
532 SECTION 2  Bacterial Diseases

FIGURE 55-1  Worldwide distribution of plague endemic areas. (Data compiled from the World Health Organization, Centers for Disease Control and Prevention, and other sources.
From Centers of Disease Control and Prevention. Prevention of plague: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 1996;45:1-15.
In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, Douglas and Bennett’s Principles and Practice of Infectious Diseases, 7th ed. Philadelphia, PA, Churchill Livingstone, Elsevier, 2011.)

wildlife species that may have died of plague. In this situation,


the organism has already produced a capsule and disseminates
more rapidly than occurs after flea-borne transmission. Organ-
isms are carried by lymphatics to regional lymph nodes. In more
than half of affected cats, this results in an intense neutrophilic
inflammatory response, variable fever, inappetence, and forma-
tion of the bubo, which is an enlarged and tender lymph node
that occasionally forms an abscess and drains purulent material.
Flea Ingestion
bite This form of disease is known as bubonic plague and is the least
fatal form of the disease. The location of the bubo(es) depends
on the initial site of infection. In cats that ingest infected rodents,
Bites, Scratches
the mandibular or retropharyngeal lymph nodes are usually
Aerosols involved, and affected cats shed bacteria from the oropharynx.
(pneumonic plague) Less often (around 20% of cats with bubonic plague), other
peripheral lymph nodes are involved; this may follow flea-borne
transmission that occurs at sites distant to the head.5
In some cats, bubo formation is accompanied by bacteremia,
Rare with associated endotoxemia and sepsis, and formation of hem-
Aerosol orrhagic, neutrophilic, and necrotizing lesions in many organs.
(pneumonic Disseminated intravascular coagulation and multiple organ
plague) failure can then ensue. Signs of severe sepsis or septic shock in
FIGURE 55-2  The role of the cat in transmission of Yersinia pestis to humans. Most affected cats include tachypnea, vomiting, tachycardia or brady-
humans are infected by direct contact with wildlife reservoir hosts or a bite from an cardia, and weak pulse quality. In some situations, bacteremia
infected flea, but infections can also occur as a result of cat bites or direct exposure to cats and multiple organ involvement occurs in the absence of identifi-
that have pneumonic plague. Dogs can also be infected but are less susceptible than cats to able buboes (septicemic form), which has a high mortality rate.
disease and shedding. Direct aerosol transmission from one human to another is rare and Pneumonic plague occurs in about 10% of affected cats as a result
only occurs in humans that have terminal pneumonic plague.
of hematogenous spread of bacteria to the lungs or, less often, by
inhalation of organisms and can result in dyspnea and cough.
72 hours on hard surfaces.8 However, Y. pestis survived at least The incubation period for plague is estimated to be 1 to 6 days,
24 days in blood-contaminated soil that was protected from UV with shorter incubation periods after ingestion or inhalation of
light.9 The extent to which environmental survival of Y. pestis the organism, and death can occur in as few as 2 to 3 days in the
contributes to the geographic distribution and epidemiology of absence of appropriate treatment; occasionally the clinical course
the disease requires further clarification. extends for 2 to 3 weeks. Some (<20%) of cats show no signs of
illness, or there may be transient fever, decreased appetite, and
Clinical Features recovery. Transient febrile illness is also common in dogs.

Signs and Their Pathogenesis Physical Examination Findings


After inoculation into the mammalian host by fleas, Y. pestis Physical examination in cats with plague reveals lethargy (82%
is taken up by mononuclear cells, in which it survives, repli- of cats), fever (71% of cats; mean rectal temperature of 105.1°F
cates, and produces F1 capsular envelope antigen. The envelope [40.5°C]), dehydration, a thin body condition, and lymphade-
allows it to further resist phagocytosis. Cats and dogs usually nopathy. Lymphadenopathy most often involves the mandib-
become infected after they ingest bacteremic rodents or other ular nodes. In one case series, 33% of 67 cats with bubonic
CHAPTER 55  Yersinia pestis (Plague) and Other Yersinioses 533

plague had bilateral lymphadenopathy, 32% had left-sided Diagnosis


lymphadenopathy, and 13% had right-sided lymphadenopathy;
in the remainder of cats, the location was not known.5 Bilat- Diagnosis of plague is based on a history of residence in an
eral disease occurred only when the mandibular and cervical endemic area as well as contact with, or predation of, wild
nodes were involved. Lymph nodes may be markedly enlarged mammals (especially rodents or lagomorphs). Duration of ill-
(up to 8 cm, with a mean of around 4 cm); may be edematous, ness, physical examination findings, and lymph node aspirate
abscessated, and painful on palpation; and may drain purulent cytology also support the diagnosis. There is a relative lack of
material. Drainage was reported in 6 (15%) of 40 cats with information available on laboratory abnormalities in dogs and
bubonic plague. Dyspnea or tachypnea can be present in cats cats with plague because the disease is rare. Ultimately, a diag-
with pulmonary involvement or possibly as a result of painful nosis of plague can only be confirmed by culture, direct fluo-
lymphadenopathy. Other reported physical examination abnor- rescent antibody testing, and/or serologic testing. Confirmation
malities in cats with plague include ocular or nasal discharge of the diagnosis may ultimately take weeks, so a high level of
(14% of cats) and abscesses that involve sites other than the suspicion for the disease is needed in endemic areas.
lymph nodes, such as the sublingual region or the tongue, oral
cavity, face, limbs, or trunk. Physical examination findings in Laboratory Abnormalities
dogs suspected to have plague have included fever, lethargy, Hematologic abnormalities in naturally infected cats most often
mandibular or cervical bubo formation, oral ulceration and consist of mild to moderate neutrophilia, usually with a mild to
ptyalism, and cough.10 moderate left shift, and occasionally lymphopenia and mono-
cytopenia.5 Mild leukopenia can occur in cats with pneumonic
plague. Platelet counts have not been widely reported in natu-
rally infected cats but may be low in cats with severe sepsis. The
serum biochemistry panel may show hypoalbuminemia, hyper-
glycemia, azotemia, electrolyte abnormalities, and increased
liver enzyme activities.11

Diagnostic Imaging
Plain Radiography
Findings on thoracic radiography in cats with pneumonic plague
have not been described in detail, but include diffuse interstitial,
alveolar, and often nodular lung opacities.

Sonographic Findings
Abdominal ultrasound findings in cats with plague have
not been reported. Based on pathologic findings, they might
FIGURE 55-3  Approximate distribution of human plague cases in western United
include abdominal lymphadenomegaly, splenomegaly, and
States, 1970 to 2012. Each dot represents a case, with the dot placed randomly in the
county of exposure. (Modified from Dennis DT, Mead PS. Yersinia species, including plague. hepatomegaly.
In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, Douglas and Bennett’s Principles and
Practice of Infectious Diseases, 7th ed. Philadelphia, PA, Churchill Livingstone, Elsevier,
Microbiologic Tests
2011. Data from 2007-2012 collated from ProMed-Mail reports, http://www.promedmail. Diagnostic assays currently available for plague in dogs and cats
org/. Last accessed February 5, 2013). are described in Table 55-1.

TABLE 55-1
Diagnostic Assays Currently Available for Plague in Dogs and Cats
Assay Specimen Type Target Performance
Culture Lymph node ­aspirates, Yersinia pestis Recent antibiotic treatment may cause false-negative results.
blood, tissues May take 48 hours for colonies to appear. Hazardous to
­collected at necropsy laboratory personnel and should be sent to laboratories with
appropriate expertise when possible.
Cytology Lymph node aspirates Y. pestis organisms Low sensitivity. Recent antibiotic treatment may cause false-
(bipolar staining bacilli) negative results.
Direct IFA Lymph node aspirates, Y. pestis F1 antigen Increases sensitivity for detection of Y. pestis over cytology
tissues collected at alone, but false negatives can still occur. Recent antibiotic
necropsy treatment may cause false-negative results. Must be per-
formed by laboratories with expertise in plague diagnosis.
Serology Serum Antibodies to Yersinia Acute and convalescent serology is required for diagnosis of acute
pestis infection, because initial results may be negative, and positive
results may reflect previous exposure rather than active infection.
534 SECTION 2  Bacterial Diseases

TABLE 55-2
Suggested Antimicrobial Drugs for Treatment of Plague in Cats
Dose Interval
Drug (mg/kg) Route (hours) Duration
Doxycycline 10 IV or PO* 24 10 days
Gentamicin 5-8 IV, IM, SC 24 10 days

*Oral administration should be avoided for at least the first 72 hours


of treatment to minimize exposure of the caregiver to Y. pestis. For IV
doxycycline administration, dilute in 100 mL of lactated Ringer’s saline
and administer over 2 hours; if fluid overload is a concern, consider
parenteral tetracycline or use of an aminoglycoside.

FIGURE 55-4  Lymph node aspirate cytology from a cat with bubonic plague. Bipolar
rods are scattered throughout the field. (From Nelson RW, Couto CG, eds. Small Animal test is a passive hemagglutination inhibition (HI) assay (see
Internal Medicine, 4 ed. St. Louis, MO: Mosby; 2009.) Chapter 2), which is available through public health laborato-
ries approved for plague diagnosis. Because of the extremely
Cytologic Demonstration of Bacteria short incubation period, a significant proportion of affected cats
Y. pestis is usually found in large numbers in tissues. If plague is and dogs may have negative titers when they first are brought
suspected, aspirates of buboes or other affected tissues should be to the veterinarian, so convalescent serum should be submitted
collected and smears prepared in isolation while wearing protec- at least 10 to 14 days after the initial titer. Some cats may die
tive clothing, including eye protection and properly fitted high- before a rise in antibody titer occurs.11 A fourfold increase in
density face masks (see Chapter 11). The smears should be heat titer together with consistent clinical abnormalities confirms the
fixed or immediately immersed in fixative solution to inactivate diagnosis of plague. Titers in affected cats in one study ranged
any organisms present. Cytologic examination shows massive from less than 1:4 for acute specimens to 1:8192 for convales-
numbers of neutrophils and a monomorphic population of coc- cent specimens.5
cobacilli with bipolar staining (Figure 55-4). This is in contrast to
cat bite abscesses, where a mixture of bacteria is usually present. Molecular Diagnosis Using the Polymerase Chain Reaction
PCR assays have been described for detection of Y. pestis DNA
Direct Immunofluorescent Antibody and Antigen ELISA Assays and are an attractive means of diagnosis because of their sen-
Heat-fixed slides prepared from lymph node aspirates can be sitivity and rapid turnaround time. In addition, the organism
submitted to approved testing facilities for application of immu- can be deliberately inactivated before specimens are submitted
nofluorescent antibody against the F1 envelope antigen. Local to the laboratory for PCR testing, which reduces the risk of
public health authorities should be contacted for information on laboratory-acquired infection. Real-time PCR assays have been
recommended testing facilities if immunofluorescent antibody validated for use on clinical specimens from humans,13 but to
(IFA) testing is required. IFA can also be applied to impression date assays have not been validated for diagnosis of the disease
smears from tissues obtained at necropsy. However, negative IFA in cats or dogs. Real-time PCR was considerably more sensitive
results do not rule out plague, so culture and serology should also for diagnosis of human plague in Africa than the lateral-flow
be considered.5 A lateral-flow dipstick assay has been described dipstick antigen assay or culture.
for rapid (within 15 minutes), sensitive, and specific in-clinic
diagnosis of plague in human patients, although false negatives Pathologic Findings
may still occur.12,13 This assay detects F1 antigen in body fluids Gross pathologic findings in cats with plague include enlarged
such as sputum, lymph node aspirates, serum, and urine and has lymph nodes or abscesses with associated edema and hemor-
an estimated shelf life of about 2 years at room temperature. It rhage, focal (up to 1 cm) lung lesions, and hemorrhages in a
has not been validated for diagnosis of plague in cats. variety of organs. Histopathology reveals hemorrhage, necrosis,
neutrophilic infiltrates, and often very large numbers of small
Isolation and Identification coccobacilli in tissues.
Y. pestis can be grown from clinical specimens on routine media
used for isolation of organisms that belong to the Enterobacte- Treatment and Prognosis
riaceae. The organism grows slowly, and colonies are smaller
than those of other Enterobacteriaceae, so cultures must be held Treatment of cats and dogs with plague involves supportive
for at least 48 hours. If plague is suspected, specimens should care, abscess drainage, and antimicrobial drug administra-
never be shipped to routine veterinary diagnostic laboratories. tion (Table 55-2). Animals suspected to have plague should
Instead, local public health authorities should be contacted be housed in isolation at least for the first 72 hours of anti-
immediately for instructions regarding how and where to sub- microbial drug therapy and handled by as few individuals as
mit specimens for culture of Y. pestis. possible. Public health authorities should be notified immedi-
ately. Gowns, gloves, and suitable face protection (eye protec-
Serology tion and a properly fited high-density surgical mask) should
Acute and convalescent serology is required for serologic diag- be worn, and protective wear should be properly removed
nosis of plague in dogs and cats. The most widely used serologic and disposed of after handling affected cats. Cats and dogs
CHAPTER 55  Yersinia pestis (Plague) and Other Yersinioses 535

Public Health Aspects


The clinical course of plague in humans closely resembles that
in cats (Figure 55-5). Three major pandemics of plague have
occurred in humans: Justinian plague, which occurred in the
Mediterranean basin during the sixth century Byzantine era; the
“Black Death,” which began in 1347 and spread throughout
Europe; and modern plague, which began in China in the 1860s
and was subsequently spread by ship rats to all continents inhab-
ited by humans. Currently, more than 95% of human plague
cases reported to the WHO originate from sub-Saharan Africa
and Madagascar.15 From 1970 to 2007, 415 cases were reported
in the United States, with 59 deaths.16 In endemic regions,
humans who have close contact with rodents or their canine
or feline predators are at risk. The vast majority of humans are
infected as a result of contact with rodent fleas. Human disease
FIGURE 55-5  Axillary bubo with ulceration and eschar formation at the site of an has also been reported after skinning of rabbits or contact with
infected flea bite. (Centers for Disease Control and Prevention.) (From Dennis DT, Mead wild carnivores. Fewer than 10% of human plague cases result
PS. Yersinia species, including plague. In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, from exposure to cats, with infection transmitted from cats to
Douglas and Bennett’s Principles and Practice of Infectious Diseases, 7 ed, Philadelphia, humans after a bite, scratch, or inhalation of respiratory droplets
PA: Churchill Livingstone, Elsevier; 2010. ) from cats with pneumonic plague.17-19 However, exposure to cats
often results in pneumonic plague, which has the worst prognosis
and can progress to death within as little as 24 hours after the
with plague should also be treated for fleas. Abscesses should onset of symptoms. Occasionally, dogs or cats carry rodent fleas
be lanced and flushed with chlorhexidine solution. Any bio- into the house, which may then transmit the infection to humans.
hazardous waste should be double bagged and disposed of Sleeping with a dog may also increase the risk of infection.20
correctly. Routine disinfectants can be used to inactivate the Exposure to infected fleas carried into a household by a dog was
organism in the environment. Antimicrobials with activity suspected in a report of two affected humans in Oregon, one of
against most isolates of Y. pestis are aminoglycosides or tetra- whom slept in the same bed with the pet dog during the 2 weeks
cyclines such as doxycycline. Trimethoprim-sulfamethoxazole before onset of illness.21 The dog seroconverted to Y. pestis.
and chloramphenicol also have activity against Y. pestis. Par- Flea control and reduction of pet exposure to rodents also
enteral aminoglycosides such as gentamicin are preferred for serve to reduce plague in humans. Pet owners should be made
seriously ill animals. Mortality rates in naturally infected cats aware that pets can carry fleas (and ticks) into the house and that
that are not treated with antimicrobial drugs can reach 75%, flea control may reduce the chance of arthropod-­transmitted
but most (>90%) of cats treated with antimicrobial drugs human disease. Adoption of wild animals should be discour-
survive.5 The time between antimicrobial drug treatment and aged. In the veterinary hospital situation, plague can be pre-
recovery of affected cats ranged from 1 to 10 days in one study vented if there is a high index of suspicion for the disease and
(mean, 4.4 days).5 appropriate precautions are taken (see Treatment and Progno-
sis). Owners of pets suspected to have plague should be advised
Immunity and Vaccination to contact their physicians without delay. Humans with expo-
sure to affected animals are generally treated with prophylactic
There are no plague vaccines for dogs and cats. Although vac- doxycycline for 7 days.
cines have been used to prevent plague in highly at-risk humans,
these did not protect against pneumonic plague. Currently there Other Yersinioses
are no vaccines approved for use in the United States. Because
Y. pestis is a potential agent of bioterrorism, efforts have been Y. pseudotuberculosis and Y. enterocolitica are enteropatho-
focused on development of subunit vaccines. However, Y. pes- genic bacteria that are distributed worldwide. They are classified
tis has also been considered an unlikely agent of bioterrorism into serotypes based on their O antigens, with different classi-
because in general it does not survive well in the environment, fication systems used for the two pathogens. Enteropathogenic
and person-to-person transmission requires close contact with a yersiniae are maintained in a variety of food animal and wild
dying person, usually on the last day of life.2,14 animal reservoirs and are well known causes of food-borne febrile
gastrointestinal illness in humans. Pigs are a major reservoir for
Prevention Y. enterocolitica, and outbreaks have followed consumption of
undercooked pork products. Y. enterocolitica can also grow in
Prevention of plague in dogs and cats involves housing cats food held at refrigeration temperatures. Affected human patients
indoors in endemic areas and discouragement of predation or frequently exhibit right lower abdominal pain due to mesenteric
exposure to/consumption of wild animal carcasses. Dogs should adenitis or ileitis, which can mimic appendicitis. A small pro-
not be allowed to dig around rodent burrows in endemic areas. portion of patients develop complications such as secondary
Flea preventatives should reduce the chance that dogs and cats polyarthritis and erythema nodosum (panniculitis).16 Children
will carry infected fleas into the house. Rodent control in the are most susceptible. Immunocompromised humans (such as
environment (e.g., removal of garbage and food scraps) should those with diabetes mellitus, malignancy, or immunosuppressive
also be performed. drug therapy) can occasionally develop bacteremia with systemic
536 SECTION 2  Bacterial Diseases

spread and abscesses in a variety of different organs. The mortal- 7. Chomel BB, Jay MT, Smith CR, et  al. Serological surveillance of
ity rate in this situation is at least 50%.16 plague in dogs and cats, California, 1979-1991. Comp Immunol
Dogs and cats can shed Y. enterocolitica and Y. pseudo- Microbiol Infect Dis. 1994;17:111-123.
tuberculosis in their feces in the absence of clinical signs.22,23 8. Rose LJ, Donlan R, Banerjee SN, et  al. Survival of Yersinia
pestis on environmental surfaces. Appl Environ Microbiol.
Y. enterocolitica was also isolated from 19% of 144 tonsils from
2003;69:2166-2171.
stray dogs in Ireland.24 Strains found in dogs can resemble those 9. Eisen RJ, Petersen JM, Higgins CL, et  al. Persistence of Yer-
implicated in human disease,25 and in several reports, dogs were sinia pestis in soil under natural conditions. Emerg Infect Dis.
suspected as a source for human infection and disease. Y. entero- 2008;14:941-943.
colitica infection was associated with hepatitis in a 2-year-old 10. Orloski KA, Eidson M. Yersinia pestis infection in three dogs. J Am
Pekingese dog from Korea26 and hepatitis and myocarditis in a Vet Med Assoc. 1995;207:316-318.
4-month-old Rottweiler.24 It has also been isolated from young 11. Gasper PW, Barnes AM, Quan TJ, et al. Plague (Yersinia pestis) in
dogs with hemorrhagic diarrhea. There have been several reports cats: description of experimentally induced disease. J Med Ento-
of Y. pseudotuberculosis infection in cats, with microabscess mol. 1993;30:20-26.
formation in a variety of abdominal organs.27-29 A young adult 12. Chanteau S, Rahalison L, Ralafiarisoa L, et al. Development and
testing of a rapid diagnostic test for bubonic and pneumonic
dog evaluated at the authors’ hospital for hemoptysis and lung
plague. Lancet. 2003;361:211-216.
consolidation had pulmonary Y. pseudotuberculosis infection. 13. Riehm JM, Rahalison L, Scholz HC, et  al. Detection of Yersinia
Enteropathogenic Yersinia species are usually susceptible to pestis using real-time PCR in patients with suspected bubonic
doxycycline, trimethoprim-sulfamethoxazole, chloramphenicol, plague. Mol Cell Probes. 2011;25:8-12.
aminoglycosides, and fluoroquinolones. Some organisms pro- 14. Kool JL. Risk of person-to-person transmission of pneumonic
duce β-lactamase enzymes. plague. Clin Infect Dis. 2005;40:1166-1172.
In dogs and cats, prevention of enteropathogenic yersiniosis 15. Stenseth NC, Atshabar BB, Begon M, et al. Plague: past, present,
involves feeding properly cooked foods and preventing preda- and future. PLoS Med. 2008;5:e3.
tion. Other measures to reduce zoonotic transmission include 16. Dennis DT, Mead PS. Yersinia species, including plague. In: Man-
hand washing after handling dogs and cats (especially before dell GL, Bennett JE, Dolin R, eds. Principles and Practice of Infec-
tious Diseases. 7th ed. Philadelphia, PA: Elsevier; 2010:2943-2953.
eating), wearing gloves when contact with saliva or feces is
17. Doll JM, Zeitz PS, Ettestad P, et  al. Cat-transmitted fatal pneu-
anticipated, and preventing dogs from licking the face or open monic plague in a person who traveled from Colorado to Arizona.
wounds. Am J Trop Med Hyg. 1994;51:109-114.
18. Thornton DJ, Tustin RC, Pienaar BJ, et  al. Cat bite transmis-
sion of Yersinia pestis infection to man. J S Afr Vet Assoc. 1975;
CASE EXAMPLE 46:165-169.
19. Werner SB, Weidmer CE, Nelson BC, et al. Primary plague pneu-
monia contracted from a domestic cat at South Lake Tahoe. Calif.
Refer to Chapter 56, Tularemia, for a case example. JAMA. 1984;251:929-931.
20. Gould LH, Pape J, Ettestad P, et al. Dog-associated risk factors for
human plague. Zoonoses Public Health. 2008;55:448-454.
SUGGESTED READINGS 21. Wilkie M, McGivern T, Skeels M, et al. Notes from the field: Two
cases of human plague—Oregon, 2010. Morb Mortal Wkly Rep.
Butler T. Plague into the 21st century. Clin Infect Dis. 2009;49:736- 2011;60:214.
742. 22. Fenwick SG, Madie P, Wilks CR. Duration of carriage and trans-
Eidson M, Thilsted JP, Rollag OJ. Clinical, clinicopathologic, and mission of Yersinia enterocolitica biotype 4, serotype 0:3 in dogs.
pathologic features of plague in cats: 119 cases (1977-1988). J Am Epidemiol Infect. 1994;113:471-477.
Vet Med Assoc. 1991;199:1191-1197. 23. Fukushima H, Nakamura R, Iitsuka S, et al. Prospective systematic
Wilkie M, McGivern T, Skeels M, et  al. Notes from the field: Two study of Yersinia spp. in dogs. J Clin Microbiol. 1984;19:616-622.
cases of human plague—Oregon, 2010. Morb Mortal Wkly Rep. 24. Murphy BP, Drummond N, Ringwood T, et al. First report: Yer-
2011;60:214. sinia enterocolitica recovered from canine tonsils. Vet Microbiol.
2010;146:336-339.
REFERENCES 25. Wang X, Cui Z, Wang H, et  al. Pathogenic strains of Yer-
1. Yersin A. La peste bubonique a Hong Kong. Ann Inst Pasteur sinia enterocolitica isolated from domestic dogs (Canis familia-
(Paris). 1894;8:662-667. ris) belonging to farmers are of the same subtype as pathogenic
2. Butler T. Plague into the 21st century. Clin Infect Dis. 2009;49: Y. enterocolitica strains isolated from humans and may be a source
736-742. of human infection in Jiangsu Province, China. J Clin Microbiol.
3. Eisen RJ, Borchert JN, Holmes JL, et  al. Early-phase transmis- 2010;48:1604-1610.
sion of Yersinia pestis by cat fleas (Ctenocephalides felis) and their 26. Byun JW, Yoon SS, Lim SK, et  al. Hepatic yersiniosis caused by
potential role as vectors in a plague-endemic region of Uganda. Am Yersinia enterocolitica 4:O3 in an adult dog. J Vet Diagn Invest.
J Trop Med Hyg. 2008;78:949-956. 2011;23:376-378.
4. Lorange EA, Race BL, Sebbane F, et al. Poor vector competence of 27. Iannibelli F, Caruso A, Castelluccio A, et al. Yersinia pseudotuber-
fleas and the evolution of hypervirulence in Yersinia pestis. J Infect culosis in a Persian cat. Vet Rec. 1991;129:103-104.
Dis. 2005;191:1907-1912. 28. Spearman JG, Hunt P, Nayar PS. Yersinia pseudotuberculosis
5. Eidson M, Thilsted JP, Rollag OJ. Clinical, clinicopathologic, and infection in a cat. Can Vet J. 1979;20:361-364.
pathologic features of plague in cats: 119 cases (1977-1988). J Am 29. Obwolo MJ, Gruffydd-Jones TJ. Yersinia pseudotuberculosis in the
Vet Med Assoc. 1991;199:1191-1197. cat. Vet Rec. 1977;100:424-425.
6. Eisen RJ, Enscore RE, Biggerstaff BJ, et al. Human plague in the
southwestern United States, 1957-2004: spatial models of ele-
vated risk of human exposure to Yersinia pestis. J Med Entomol.
2007;44:530-537.
CHAPTER 56

Tularemia
Jane E. Sykes and Bruno B. Chomel

F. tularensis subsp. tularensis has the greatest degree of viru-


Overview of Tularemia lence for humans and predominates in North America, whereas the
First Described: 1911, Tulare County, California1 less virulent subspecies holarctica predominates in Europe. Addi-
Cause: Virulent strains of Francisella tularensis, a gram-­ tional genetic variation within type A and type B subpopulations
negative coccobacillus (family Francisellaceae) of F. tularensis correlates with geographic distribution of cases and
the severity of disease.2 Molecular subtyping has further divided
Major Vectors: Ticks (especially Dermacentor andersoni, Der-
type A into subpopulations A1a, A1b, A2a, and A2b. The highest
macentor variabilis, and Amblyomma americanum in the
mortality in humans, 24%, results from infections by A1b.3
eastern United States), biting flies (western United States),
Tularemia occurs primarily in the Northern Hemisphere, espe-
mosquitos (Scandinavia)
cially between 30 degrees and 71 degrees of latitude (Figure 56-1).
Affected Hosts: Some 190 mammalian species, which include Human disease caused by a subspecies novicida–like organism
rodents, rabbits, wild carnivores, domestic dogs and cats, was reported from the Northern Territory in Australia in 2003.4
sheep, horses, nonhuman primates, and humans Even in North America, where F. tularensis subsp. tularensis
Geographic Distribution: Northern Hemisphere between predominates, tularemia is a rare disease, and most human cases
30 degrees and 71 degrees of latitude; most disease is occur in the central and north-central United States, especially
reported from the United States, where virulent strains Arkansas, Missouri, Kansas, South Dakota, and Oklahoma, with
are most prevalent an additional focus in Massachusetts (especially Martha’s Vine-
yard) (Figure 56-2). Despite its rarity, tularemia is a reportable
Major Clinical Signs: Fever, lethargy, inappetence, lymph-
disease in the United States because F. tularensis is considered a
adenopathy, subcutaneous abscesses, splenomegaly,
potential agent of bioterrorism. In western Europe, tularemia is
hepatomegaly
endemic in Scandinavia but has emerged in new parts of Sweden
Differential Diagnoses: Plague, cat bite abscess, Streptococcus and is increasingly reported in humans from more southern loca-
spp. infections, feline infectious peritonitis, mycobacte- tions, such as Germany, France, and Spain.5 Tularemia has been
rial infections, lymphoma, sepsis caused by other gram- absent from the United Kingdom, with the exception of travel-
negative bacteria related disease. It also occurs in Russia and Japan.6
Human Health Significance: Cats and less commonly dogs Canine and feline disease due to F. tularensis has only been
can transmit tularemia to humans after they are exposed reported from the United States. Young adult cats and dogs are
to infected rodents or rabbits. Cat bites are most com- usually affected. As with plague, cats are more susceptible than
monly implicated. dogs. Cases are reported most often from Oklahoma7-9 and
Kansas,10 but also from other locations that include Montana,11
Missouri,12 Massachusetts,13 central Virginia,14 and eastern
Oregon,15 which mirrors the distribution in humans. Tularemia
is likely underdiagnosed in cats and dogs because clinical signs
Etiologic Agent and Epidemiology
Tularemia (also known as rabbit fever, hare fever, deerfly fever,
and lemming fever) is an extremely infectious but uncommon
zoonotic disease caused by Francisella tularensis, a small (0.2
× 0.2 × 0.7 µm), nonmotile, aerobic, encapsulated gram-nega-
tive coccobacillus. The name “tularemia” comes from the first
isolation of the organism from sick rodents in Tulare County,
California, in 1911. There are four subspecies of F. tularen-
sis: tularensis, holarctica, novicida, and mediasiatica. Subspe-
cies tularensis (also known as type A) and holarctica (type B)
cause most disease in humans. Subspecies mediasiatica is only
found in Asia and the former Soviet Union. Subspecies novicida
can cause illness in severely immunosuppressed humans and is
widely studied as a model for tularemia because of its low viru- FIGURE 56-1  Global distribution of tularemia (Centers for Disease Control and
lence to laboratory workers. Prevention).

537
538 SECTION 2  Bacterial Diseases

Ingestion of dead rabbit


by dogs or cats

Bite wound
(usually cat)

Arthropod

biting fly
A
ticks (esp. Dermacentor spp.)
Terrestrial or
aquatic
reservoirs

Inhalation

Ingestion
Cutaneous inoculation
FIGURE 56-3  Transmission cycle for Francisella tularensis.

are nonspecific. Many cats with tularemia are diagnosed with


tularemia at necropsy.7,10 The seroprevalence in pet cats from
Connecticut and New York state was 12%.16
F. tularensis is maintained in the environment by a huge
variety of terrestrial and aquatic mammals, which include rab-
Type A Human Type B Human bits, hares, ground squirrels, prairie dogs, lemmings, voles,
B Type A Animal Type B Animal beavers, muskrats, water rats, and other rodents.6 It often
causes severe disease in these species, although subclinical car-
riage may also occur. It can be transmitted by arthropod bites,
inhalation or ingestion of the organism, and direct skin contact
with infected tissues (probably when there are minute breaks
in the skin) (Figure 56-3). Inhalation of as few as 10 organisms
can cause human disease, whereas larger numbers of organ-
isms (108) must be ingested for disease to occur. Cats and dogs
become infected when they hunt rodents or lagomorphs; a his-
tory of rabbit ingestion was most often described in association
with the few reported feline and canine cases.7-9 In the western
United States (Utah, Nevada, and California), the most com-
mon arthropod vectors are biting flies, whereas ticks (especially
Dermacentor ticks and Amblyomma americanum) are the most
common vectors east of the Rocky Mountains. In Europe, Der-
macentor ticks and Ixodes ricinus are the main tick vectors;
Type A1a Human Type A1b Human Type A2 Human
mosquitos are important vectors in Scandinavia.17 F. tularensis
is well known for its ability to survive up to several months
Type A1a Animal Type A1b Animal Type A2 Animal
in fresh water sources, and survival in brackish water has also
C been demonstrated.18 Survival and replication of F. tularensis
FIGURE 56-2  Geographic distribution of tularemia in the USA. Compare this with within free-living amoebae such as Hartmannella vermiformis
the geographic distribution of plague (Figure 55-3). A, Overall distribution of human may contribute to the organism’s ability to persist in water
tularemia cases. B, Distribution of type A and type B isolates from humans and animals. sources.19 The organism can also survive for years in frozen
C, Distribution of isolates from humans and animals by molecular subtype. Note that the rabbit meat.
geographic distribution of human type A cases follows the distribution of subtype A1, the
most virulent subtype being A1b. (A and B modified from Kugeler KJ, Mead PS, Janusz AM, Clinical Features
et al. Molecular epidemiology of Francisella tularensis in the United States. Clin Infect Dis
2009;48:863-870.) Signs and Their Pathogenesis
The clinical manifestations of tularemia depend on the virulence
of the infecting strain, the route of inoculation, and host immu-
nity. Although tularemia may rapidly progress to death in cats,
protracted illness has also been described.15
CHAPTER 56  Tularemia 539

F. tularensis survives in macrophages through evasion of biosafety level 3 procedures is required to perform these tests
cellular destruction by phagosomes and inhibition of the respi- when tularemia is suspected. Local public health authorities
ratory burst. Several different virulence factors of F. tularensis should be contacted to report suspicion for the disease and for
have been identified, which include bacterial lipopolysaccharide information on recommended testing facilities.
(LPS), pili, a capsule, and a series of acid phosphatase enzymes.
The endotoxic activity of the LPS of F. tularensis is 1000-fold Laboratory Abnormalities
less potent than that of Escherichia coli.20,21 Production of acid Complete Blood Count, Biochemistry Profile, and Urinalysis
phosphatase enzymes may be key to the organism’s ability to Leukocyte counts in a limited number of cats with tularemia have
inhibit the respiratory burst and survive in macrophages.22 been normal, decreased (as low as 1300 cells/µL), or increased
Many stages of the organism’s intracellular lifestyle depend on with evidence of neutrophil toxicity.7,9,13,14 Moderate to marked
a pathogenicity island (group of virulence genes) known as the bandemia may be present.9,13 Thrombocytopenia is a common
Francisella pathogenicity island. finding (as low as 41,000 platelets/µL).7,9 The serum biochem-
For the first few days after cutaneous inoculation, F. tula- istry panel may show hypoglycemia, azotemia, and electrolyte
rensis replicates locally and can produce a papule that may abnormalities, increased liver enzyme activities, and hyperbiliru-
then ulcerate. It then disseminates to local lymph nodes, which binemia (up to 7.6 mg/dL).7,9,14 Bilirubinuria and hemoglobin-
may be followed by bacteremia and sepsis, with infection of uria may be present.7 Mature neutrophilia, lymphopenia, and
systemic, primarily reticuloendothelial tissues. The organism increased serum ALP activity were present in a dog with tula-
invades not only macrophages but also nonphagocytic cells remia.8 In human patients, abnormalities include thrombocyto-
such as hepatocytes and alveolar epithelial cells,23 with induc- penia, elevated serum transaminase activities, increased serum
tion of a profound inflammatory response and necrosis. Incuba- creatine kinase activity, myoglobinuria, and pyuria.26
tion periods that range from 1 to 5 days have been suspected in
naturally occurring tularemia in dogs and cats, but longer incu- Cytologic Examination of Lymph Node Aspirates
bation periods (up to 21 days) can occur in humans. Clinical Cytologic examination of lymph node aspirates in cats with
signs typically manifest as a sudden onset of fever, lethargy, and tularemia may reveal lymph node hyperplasia.9 Lymph nodes
inappetence. Other manifestations in affected cats have included may also contain necrotic debris and increased numbers of mac-
a thin body condition, icterus, hepatomegaly, vomiting, dehy- rophages and/or degenerate neutrophils.7,13 Because F. tularen-
dration, peripheral and/or abdominal lymphadenomegaly, sple- sis is tiny and stains only faintly, organisms are less likely to be
nomegaly, and oral and/or lingual ulceration. Hypothermia and visualized than those of Yersinia pestis (see Chapter 55).
bradycardia can occur in moribund cats7; one cat had seizures
and was nonresponsive.14 A single cat with localized cutaneous Diagnostic Imaging
tularemia had a chronic (1-year duration) draining skin lesion in Plain Radiography
the region of (but not involving) the mandibular lymph nodes, Findings on thoracic radiography in cats with tularemia have
in the absence of systemic signs.15 In the rare reports of natu- not been well described; one cat had normal thoracic radio-
rally affected dogs, clinical signs have included fever, anorexia, graphs.9 Findings on abdominal radiography have included
and peripheral lymphadenopathy.8,24 One dog also had bilateral hepatomegaly and fluid-filled intestinal loops.9
mucoid ocular discharge and mild conjunctival hyperemia,8 and
another had tonsillitis.24 Sonographic Findings
Abdominal ultrasound findings in cats with tularemia could
Physical Examination Findings include abdominal lymphadenomegaly, splenomegaly, and/or
Physical examination in cats with tularemia may reveal lethargy, hepatomegaly. Abdominal lymphadenomegaly was described in
fever (up to 105.1°F [40.5°C]) or hypothermia, a thin body con- one cat.9
dition, and dehydration.7,9,11-13 Ulceration of the oral mucous
membranes or tongue may be present.7,13 Icterus may also Microbiologic Tests
occur.25 Moribund cats may be hypothermic and bradycardic and Diagnostic assays currently available for tularemia in dogs and
may have weak pulse quality. Mild to marked peripheral lymph- cats are described in Table 56-2.
adenopathy is present in most but not all cats. Lymphadenopathy
may be restricted to the lymph nodes of the head and neck13 or Direct Detection
may be generalized.7,9 Abdominal palpation may reveal hepa- Heat-fixed slides can be submitted to experienced laborato-
tomegaly or splenomegaly, or abdominal lymph nodes may be ries for direct immunofluorescent antibody (IFA) staining for
palpable. Fever, lethargy, peripheral lymphadenopathy, mucoid F. tularensis. Alternatively, IFA or immunoperoxidase stains can
ocular discharge, and tonsillitis may be present in dogs.8,24 be applied to tissues obtained at necropsy.10 Negative results
do not rule out tularemia, but in some cases IFA may detect
Diagnosis the organism when culture is negative.27 Fluorescence in situ
hybridization (FISH) has also been used to detect F. tularensis
Tularemia should be considered in cats and dogs with lymphade- in tissues and differentiate between subspecies.28 As for plague,
nopathy and fever that have a history of residence in or recent lateral-flow assays have been described for rapid detection of
travel to an endemic area, as well as contact with or predation F. tularensis LPS antigen in clinical specimens such as urine.29
of wild mammals (especially lagomorphs). Plague is the major The use of these assays in dogs and cats has not been described.
differential diagnosis of zoonotic importance in the southwest-
ern United States (Table 56-1). Ultimately, the diagnosis can be Isolation and Identification
confirmed by culture, PCR, and/or serologic testing. With the F. tularensis can be grown from clinical specimens, but it is very
exception of PCR, access to approved laboratories that practice fastidious, grows slowly, is hazardous to laboratory personnel,
540 SECTION 2  Bacterial Diseases

TABLE 56-1
Differences and Similarities between Plague and Tularemia in Cats and Dogs in the United States
Plague Tularemia
Organism Gram-negative bacillus (Enterobacteriaceae). Gram-negative coccobacillus (Francisellaceae). Can
­Survives relatively poorly in the environment. persist in the environment and water sources
Grows on routine culture media used for gram- for months. Fastidious growth in culture; often
negative bacteria. requires special media and prolonged incubation.
Major Arthropod Diverse variety of rodent and rabbit fleas Dermacentor spp. and Amblyomma americanum
vectors ticks
Geographic location Southwestern United States Western, southwestern, central, and eastern United
States
Affected hosts Cats > dogs Cats > dogs
Usual history Rodent contact or exposure to rodent fleas Rodent and especially rabbit contact; ingestion
often reported
Clinical signs Acute disease with high fever and lymphadenopathy, Acute or more chronic disease (sometimes associ-
most often of the mandibular and/or retropharyn- ated with a thin body condition) with oral ulcer-
geal lymph nodes. Hyperbilirubinemia and icterus ation, lymphadenopathy of the mandibular and/
do not appear to be common in affected cats. or retropharyngeal lymph nodes, or generalized
lymphadenopathy and abdominal organomegaly.
Hyperbilirubinemia is frequent in affected cats,
and icterus has been described.
Lymph node aspirate Neutrophilic inflammation; monomorphic popula- Lymphoid reactivity or neutrophilic to pyogranulo-
cytology tion of bipolar staining bacilli often present matous inflammation and necrosis; bacteria often
not visualized
Antimicrobial drugs Aminoglycosides, doxycycline, chloramphenicol, Aminoglycosides are the treatment of choice. Doxy-
trimethoprim-sulfamethoxazole cycline, chloramphenicol, and fluoroquinolones
may be effective but may be followed by relapse.
Zoonotic potential Exposure to cats with pneumonic plague can lead Most often follows bites from affected cats, less of-
to human infection; bites and scratches have also ten scratches. Course of disease longer and mor-
been implicated. Human pneumonic plague usu- tality may be lower than occurs with pneumonic
ally ­results from exposure to infected cats and can plague. Medical attention should be sought if
progress to death within a few days. Exposed hu- bites or scratches occur.
mans may require prophylaxis with doxycycline.

TABLE 56-2
Diagnostic Assays Currently Available for Tularemia in Dogs and Cats
Assay Specimen Type Target Performance
Culture Lymph node aspirates, Francisella tularensis Recent antibiotic treatment may cause false-­
blood, tissues ­collected negative results. May require special media and
at necropsy takes at least 48 hours for colonies to appear.
Hazardous to laboratory personnel and should
be sent to laboratories with appropriate expertise
when possible.
Direct immunofluorescent Lymph node aspirates, F. tularensis antigen May be positive when culture is negative. False
antibody tissues collected at negatives can still occur. Recent antibiotic
necropsy ­treatment may cause false-negative results.
Serology Serum Antibodies to Acute and convalescent serology is required
F. tularensis for diagnosis, because initial results may
be ­negative and positive results may reflect
­previous ­exposure rather than active infection.
­Seroconversion may take up to 3 weeks.
CHAPTER 56  Tularemia 541

and does not always grow on media used for routine isolation
of gram-negative bacteria such as MacConkey agar (in contrast TABLE 56-3
to Y. pestis). The use of cysteine-supplemented media (such as Suggested Antimicrobial Drugs for Treatment of Tularemia in
cysteine heart agar) and incubation in a CO2-supplemented
Cats*
environment enhances recovery of the organism. Growth gener-
ally occurs 48 to 72 hours after inoculation, but cultures must
Dose Interval
be held for 7 to 10 days. Overgrowth of contaminating bacteria
Drug (mg/kg) Route (hours) Duration
can occur unless the media is supplemented with antibiotics that
inactivate these organisms. F. tularensis may grow from blood Gentamicin 5-8 IV, IM, SC 24 14 days
specimens after inoculation of routine blood culture media. The Doxycycline 5 PO† 12 2-3 weeks
organism is identified on the basis of biochemical testing with Marbofloxacin 2.75-5.5 PO 24 2-3 weeks
or without the use of molecular methods such as 23S rRNA
*The same antimicrobial drugs could be used to treat dogs; doses shown
gene PCR sequencing (which allow subspecies determination).
are for cats.
Whole-cell matrix-assisted laser desorption ionization–time of †Parenteral aminoglycosides are preferred for at least the first 72 hours
flight (MALDI-TOF) mass spectrometry, which measures the of treatment to minimize exposure of the caregiver to F. tularensis dur-
mass of peptides and small proteins from whole cells, has also ing treatment.
been used for identification of F. tularensis isolates to the sub-
species level.30
Treatment and Prognosis
Serology
As for plague, acute, and convalescent serology, with a fourfold Treatment of cats (and, although less commonly affected, dogs)
increase in titer, is required for serodiagnosis of tularemia in with tularemia involves supportive care and antimicrobial drug
affected dogs and cats and is less hazardous to human health administration (Table 56-3). As for plague, animals suspected to
than culture. Tube agglutination and microagglutination assays have tularemia should be housed in isolation at least for the first
are the standard methods for serodiagnosis, but ELISA assays 72 hours of antimicrobial drug therapy and handled by as few
have also been developed. Because of the extremely short incu- individuals as possible. Public health authorities should be noti-
bation period, affected cats and dogs may have negative titers fied. Gowns, gloves, and suitable face protection (eye protec-
when they first are brought to the veterinarian, so convalescent tion and a properly fitted high density surgical mask) should be
serum should be submitted at least 2 to 3 weeks after the initial worn, and protective wear should be properly removed and dis-
titer if the animal survives. Some cats die from the disease before posed of after handling affected cats. Any biohazardous waste
a rise in antibody titer occurs. In addition, high titers may per- should be double bagged and disposed of correctly. Routine dis-
sist after recovery from infection, so a single positive titer is not infectants should be used to inactivate the organism in the envi-
diagnostic for tularemia. ronment. The drug of choice for initial treatment of tularemia
is a parenteral aminoglycoside such as gentamicin. Doxycycline
Molecular Diagnosis Using the Polymerase Chain Reaction can also be used but is associated with a higher relapse rate in
PCR assays that detect F. tularensis DNA represent an attractive humans than occurs with aminoglycosides.26 Fluoroquinolones
means of diagnosis because of their sensitivity, rapid turnaround are active against F. tularensis in  vitro, but like tetracyclines,
time, low risk to laboratory personnel, and the ability of some they may be associated with relapse in vivo.32 Extension of the
assays to differentiate between subspecies and genotypes of F. tula- course of treatment from 2 weeks to 3 weeks when fluoroquino-
rensis.31 A PCR assay was used to detect F. tularensis in tissues at lones or doxycycline is used has been effective for treatment of
necropsy in a cat.12 A number of real-time PCR assays have been human tularemia.25 Other drugs with in vitro activity against F.
used to detect F. tularensis in human clinical specimens. Despite tularensis include chloramphenicol and erythromycin.33
the sensitivity of PCR assays, false negatives can still occur. Real-
time PCR assays for F. tularensis are offered on a commercial basis Immunity and Vaccination
by some veterinary diagnostic laboratories in the United States.
Natural infection by F. tularensis is followed by protection
Pathologic Findings against reinfection. Because F. tularensis is facultatively intracel-
Gross pathologic findings in cats with tularemia include icterus, lular, cell-mediated immune responses are required for patho-
oral ulceration, lymphadenomegaly, splenomegaly, and hepa- gen clearance, although humoral immunity is also important.23
tomegaly. Multifocal to coalescing irregular yellow, gray, or As a result, inactivated vaccines have not been effective for pre-
white foci (usually 1 to 5 mm in diameter) are present in these vention of tularemia, because they primarily induce humoral
organs, and sometimes other organs such as the lung and myo- immunity. An attenuated live vaccine that contained F. tular-
cardium.7,12,13 When examined histopathologically, the nodules ensis subsp. holarctica (live vaccine strain, or LVS) has been
represent well-circumscribed but non-encapsulated masses of available for prevention of serious disease in people at high risk
caseous necrosis, with abundant cellular debris, surrounded of exposure. However, this remains unlicensed because of con-
by infiltrates of degenerate neutrophils, macrophages, and/or cerns regarding efficacy and the route of administration, which
fibroblasts. Lesions are centered on the splenic white pulp and is by scarification.34 Since then there has been strong interest
cortical lymphoid follicles within lymph nodes. Necrosis and in vaccine development for prevention of human tularemia,
hemorrhage of lymphoid follicles within the gastrointestinal because of concerns that the organism will be used as a weapon
tract may be present.7,11 Organisms may be discernible with sil- of bioterrorism.23 Vaccines are not likely to be available for
ver staining as tiny coccobacilli11 or can be demonstrated with dogs and cats given the rarity of disease in these species and the
immunoperoxidase or immunofluorescence methods.10,27 minor role that they play in transmission of disease to humans.
542 SECTION 2  Bacterial Diseases

Prevention
Prevention of tularemia in dogs and cats involves housing cats
indoors and discouragement of predation or exposure to dead
wild rodents and rabbits. Use of tick preventatives also reduces
the chance that dogs and cats will carry infected ticks into the
house. Rodent control in the environment (e.g., removal of
­garbage) should also be performed.

Public Health Aspects


Humans are most commonly infected by F. tularensis after
arthropod bites or handling infected wild animals. In the United
States, disease is most commonly reported in late spring and
summer. Occupations associated with an increased risk of tula- FIGURE 56-4  Papule undergoing central necrosis with desquamation on the thigh
remia include farming, hunting, landscaping, meat handling, of a middle-aged man infected with Francisella tularensis. (Courtesy of Dr. Joseph A. Boc-
working with sheep, veterinary practice, and laboratory work. chini, Louisiana State University Health Sciences Center, Shreveport, LA. In: Mandell GL,
Severe illness is most often reported in young children (5 to Bennett JE, Dolin R, eds. Principles and Practice of Infectious Diseases, 7th ed, Philadelphia,
9 years) and the aged (>75 years). Men are more likely to be PA: Churchill Livingstone; 2010:2927-2937.)
affected than women.26
Several overlapping forms of disease have been described by fever and severe throat pain; tonsillitis, oral ulceration, and
in humans, the development of which depends on the route of associated lymphadenopathy may be present, which resembles
inoculation and the virulence of the infecting strain: ulceroglan- the disease described in dogs and cats (which usually acquire
dular, glandular, oculoglandular, oropharyngeal, typhoidal, and infection via ingestion). Oropharyngeal tularemia tends to
pneumonic.26 Ulceroglandular tularemia results from cutaneous occur in children who ingest contaminated food or water and
inoculation, oropharyngeal disease follows ingestion and pneu- can resemble “strep throat.”26 Typhoidal tularemia is a febrile
monic tularemia can result from aerosol inhalation. Pneumonic illness in the absence of prominent lymphadenopathy and may
tularemia has been described in people after activities such as be associated with sore throat, abdominal pain, diarrhea, vom-
lawn mowing or brush cutting in tick-infested areas. Infection iting, icterus, and, with more chronic illness, hepatomegaly and
of humans from dogs and cats can occur after these animals splenomegaly. Pneumonic tularemia is the most severe form
carry F. tularensis in their mouths after ingestion of wild prey. and results from inhalation of the organism or hematogenous
Human-to-human transmission does not occur. spread of the organism to the lung, with clinical signs such as
Clinical signs in humans consist of acute onset of a flu-like cough, chest pain, and hemoptysis. Thoracic radiographs can
illness, with fever, chills, and malaise. Sore throat, abdomi- reveal lobar infiltrates, hilar lymphadenopathy, pleural effusion,
nal pain, vomiting, diarrhea, lymphadenopathy, and a cough and miliary infiltrates.26 Changes resemble those seen in other
may also occur. Infection with less virulent strains may result chronic pneumonias, such as tuberculosis or fungal disease.
in transient flu-like illness followed by recovery. Without treat- Although not the most common route of infection for humans,
ment, clinical signs can persist for several months, with chronic cat-bite–associated tularemia has been described (including
lymphadenopathy, weight loss, and debilitation.26 Disease in among veterinary staff), and cats that live in rural endemic areas
chronically affected humans can resemble cat scratch disease. are important vectors of the most virulent strain of F. tular-
Ulceroglandular tularemia is the most common form of tula- ensis, type A1b.25 Disease in humans can mimic Pasteurella
remia seen in humans. It consists of a red and painful papule multocida cellulitis, the most common bacterial infection after
at the site of skin infection, with associated lymphadenopathy a cat bite, but treatment with amoxicillin or clavulanic acid–
(Figure 56-4). The papule then undergoes necrosis, forming amoxicillin is ineffective. Less commonly, human disease fol-
an ulcer, which can take weeks to heal. In some individuals, lows cat scratches or casual contact with cats.27 In some cases,
lymph nodes may suppurate and become fluctuant. Multiple cats had recent contact with or ingested wild rabbits or rodents.
lesions can occur on the hands when infected animals have Both apparently healthy and sick cats have been involved. Aero-
been handled, whereas tick bites typically result in single lesions sol exposure to organisms on the coats of dogs (such as occurs
elsewhere on the body.26 Only lymphadenopathy may be pres- when dogs shake their coats after contact with infected rabbits)
ent if the ulcer heals before the time of evaluation (glandular has rarely been suspected in clusters of human disease.35-37 In
tularemia). Oculoglandular tularemia occurs when organisms summary, tularemia should especially be considered in humans
gain access through the conjunctiva and is characterized by con- who develop illness in endemic areas within 3 weeks after a bite
junctivitis, conjunctival ulceration, lacrimation, and associated from a cat that is a stray or that has had a recent history of wild
lymphadenopathy.26 Oropharyngeal tularemia is characterized rabbit or rodent contact.
CHAPTER 56  Tularemia 543

CASE EXAMPLE Respiratory: Tachypnea was present with a shallow respiratory


pattern. Normal bronchovesicular sounds were present in all
lung fields.
Signalment: “Sam,” a 5-year-old male neutered domestic Abdominal Palpation: No clinically significant abnormalities
shorthair cat from Woodland in northern California were identified. The urinary bladder was palpable but
History: Sam was brought to the University of California, small.
Davis, veterinary emergency service for evaluation of fever Lymph Nodes: The left mandibular lymph node was enlarged
and inappetence. The owner first noticed he was ill 4 days at 2 cm. All other lymph nodes appeared to be within normal
previously, when he regurgitated a small amount of pink limits.
fluid about 1 hour after eating. The following day he was Laboratory Findings:
not eating and lethargic, and his water consumption was CBC:
reduced. On day 3, he was taken to a local veterinary clinic HCT 20.3% (30%-50%)
where he was found to be febrile (105°F [40.5°C]), icteric, and MCV 42.9 fL (42-53 fL)
had a mass on the left side of his neck. He was treated with MCHC 33.0 g/dL (30-33.5 g/dL)
enrofloxacin (4.5 mg/kg SC, once), ketoprofen (1.9 mg/kg Reticulocytes 9500 cells/µL (7000-60,000 cells/µL)
SC), and clavulanic acid–amoxicillin (12 mg/kg PO q12h). WBC 23,120 cells/µL (4500-14,000 cells/µL)
The results of a CBC showed a mild neutrophilia (13,083 Neutrophils 18,958 cells/µL (2000-9000 cells/µL)
cells/µL; reference range, 2500-12,500 cells/µL) with slightly Band neutrophils 3699 cells/µL
toxic neutrophils and a mild lymphopenia (1176 cells/µL; Lymphocytes 231 cells/µL (1000-7000 cells/µL)
reference range, 1500-7000 cells/µL). A biochemistry panel Monocytes 231 cells/µL (50-600 cells/µL)
showed hyponatremia (133 mEq/L; reference range, 147- Eosinophils 0 cells/µL (150-1100 cells/µL)
156 mEq/L), hypochloremia (99 mEq/L; reference range, Basophils 0 cells/µL (0-50 cells/µL)
111-125 mEq/L), hypokalemia (3.4 mEq/L; reference range, Platelets 102,000/µL (180,000-500,000 platelets/µL).
3.9-5.3 mEq/L), hyperglycemia (214 mg/dL; reference range, Serum Chemistry Profile:
70-150 mg/dL), hyperbilirubinemia (3.7 mg/dL; reference Anion gap 18 mmol/L (13-27 mmol/L)
range, 0-0.4 mg/dL), increased serum CK activity (1858 U/L; Sodium 143 mmol/L (151-158 mmol/L)
reference range, 64-440 U/L), and a mild metabolic acidosis Potassium 3.2 mmol/L (3.6-4.9 mmol/L)
(bicarbonate 12 mEq/L; reference range, 13-25 mEq/L). Chloride 110 mmol/L (117-126 mmol/L)
ELISA serology for FeLV antigen and FIV antibody and IFA Bicarbonate 18 mmol/L (15-21 mmol/L)
serology for FCoV antibody were negative. On receipt of the Phosphorus 3.0 mg/dL (3.2-6.3 mg/dL)
laboratory test results, the local veterinarian recommended Calcium 8.8 mg/dL (9.0-10.9 mg/dL)
referral. BUN 15 mg/dL (18-33 mg/dL)
Sam had been fed commercial dry and canned cat food and Creatinine 0.8 mg/dL (1.1-2.2 mg/dL)
was treated monthly with fipronil. He had been acquired at Glucose 151 mg/dL (63-118 mg/dL)
6 weeks of age, at which time he was neutered. He had been Total protein 6.6 g/dL (6.6-8.4 g/dL)
vaccinated annually by the owner for feline respiratory vi- Albumin 2.5 g/dL (2.2-4.6 g/dL)
ruses and feline panleukopenia virus, and was vaccinated for Globulin 4.1 g/dL (2.8-5.4 g/dL)
rabies for the first time 4 months earlier. He lived with three ALT 30 U/L (27-101 U/L)
other cats, three dogs, and two rabbits, and the owner had AST 39 U/L (17-58 U/L)
brought home a wild mouse from the Lake Tahoe (Sierras) re- ALP 5 U/L (14-71 U/L)
gion several weeks before Sam became ill. The rabbits were GGT < 3 U/L (0-4 U/L)
acquired by the owner 6 months ago, were kept indoors, and Cholesterol 134 mg/dL (89-258 mg/dL)
did not receive flea control. The owner fostered cats and Total bilirubin 2.1 mg/dL (0-0.2 mg/dL)
occasionally dogs for the local animal shelter and cared for Magnesium 2.0 mg/dL (1.5-2.5 mg/dL)
a colony of feral cats, which did not interact with the house- Creatine kinase 586 U/L (73-260 U/L).
hold cats. The household cats lived harmoniously indoors. Urinalysis: SGr 1.014; pH 8.0, trace protein (SSA), 1+ bilirubin,
Physical Examination: 2+ hemoprotein, 2+ glucose, rare WBC/HPF, 7-12 RBC/HPF,
Body Weight: 5.4 kg few lipid droplets.
General: Lethargic but responsive. Resented handling. T = Imaging Findings:
105.9°F [41.1°C], RR = 60 breaths/min, HR = 176 beats/min, Thoracic Radiographs and Abdominal Ultrasound: No
CRT <2 s. clinically significant abnormalities were present.
Integument: Mild scaling was present throughout the haircoat, Cervical Ultrasound: There was moderate to severe
but no ectoparasites were visible. enlargement of the retropharyngeal lymph nodes bilaterally
Eyes, Ears, Nose, and Throat: No clinically significant and of the left mandibular salivary gland.
abnormalities were present. Microbiologic Testing: Cytologic examination of lymph
Musculoskeletal: Body condition score 6/9. There was a 2 × 2 node aspirates (mandibular and retropharyngeal): A
× 6 cm firm subcutaneous mass alongside the trachea on slightly bloody background was admixed with markedly
the left side. increased numbers of variably degenerate neutrophils.
Cardiovascular: There were no murmurs or arrhythmias Low numbers of lymphocytes and scattered macrophages
detected on cardiac auscultation. Pulses were strong and are also observed. Interpretation: marked suppurative
synchronous. inflammation.

Continued
544 SECTION 2  Bacterial Diseases

Comments: Although no intracellular bacteria were seen, the Comments: A diagnosis of plague or tularemia was never
number of neutrophils and their variably degenerate nature confirmed in this cat, although strongly suspected based
suggested sepsis as the primary differential diagnosis. on the history of wild rodent and rabbit exposure (with
Aerobic and anaerobic bacterial culture and susceptibility the rodent adopted from a plague endemic region), and
(lymph node aspirate): No growth. the clinical abnormalities that were present. The findings
PCR for Francisella tularensis DNA (lymph node aspirate): Negative. of hyperbilirubinemia, thrombocytopenia, and increased
Diagnosis: Severe suppurative lymphadenitis and fever; serum CK activity were perhaps more suggestive of
suspect plague or tularemia. tularemia than plague, although hyperbilirubinemia and
Treatment: Sam was transferred to isolation, during which thrombocytopenia can occur in human plague. The history
time he bit a clinician, and treated with intravenous fluids of antibiotic treatment may have contributed to the negative
(lactated Ringer’s solution and 20 mEq/L KCl at 15 mL/h IV) cytology, culture, and PCR results. Culture was requested on
and antimicrobial drugs (ampicillin, 20 mg/kg IV q8h and site shortly after admission because plague and tularemia
gentamicin, 5 mg/kg IV q24h). Public health officials were were initially overlooked as likely diagnoses. Once these
contacted. The cat’s rectal temperature initially increased diseases were considered, an attempt was made to send
to 107°F (41.6°C); dark brown diarrhea was also noted. On specimens out to a public health laboratory for additional
the second day of hospitalization, Sam’s rectal temperature testing, but this was complicated by concerns that related to
was 105°F (40.5°C), and by day 3 it had normalized and the potentially hazardous nature of the specimens. Handling
Sam began to eat. It increased again to 103.9°F (40.0°C) the of the cat was limited to as few individuals as possible and
following day, and the right mandibular and retropharyngeal was difficult because the cat was aggressive. The clinician
lymph nodes became palpably enlarged. Subsequently, who was bitten by the cat immediately saw her physician,
the cat’s rectal temperature normalized, laboratory who prescribed doxycycline for 7 days. Rabies quarantine
abnormalities improved, and the lymph nodes decreased was also instituted because the cat was incompletely
in size and were soft on palpation. The cat was discharged vaccinated for rabies. The owner was educated about the
with instructions to continue treatment with oral amoxicillin possibility of plague or tularemia and the implications for
and subcutaneous gentamicin for 7 days, with a recheck human health. Although flea control was used, not all of the
planned at that time for an examination and convalescent Y. animals received flea prevention, and there was significant
pestis and F. tularensis serology. The owner did not bring Sam turnover of a variety of wildlife and domestic animal species
back for a recheck examination, but a follow-up telephone in the household. Continued adoption of wild animals by the
conversation indicated that Sam had recovered completely. owner was discouraged.

SUGGESTED READINGS 9. Woods JP, Crystal MA, Morton RJ, et al. Tularemia in two cats.
J Am Vet Med Assoc. 1998;212:81-83.
Kugeler KJ, Mead PS, Janusz AM, et  al. Molecular epidemiology of 10. DeBey BM, Andrews GA, Chard-Bergstrom C, et al. Immunohisto-
Francisella tularensis in the United States. Clin Infect Dis. 2009;48: chemical demonstration of Francisella tularensis in lesions of cats
863-870. with tularemia. J Vet Diagn Invest. 2002;14:162-164.
Spagnoli ST, Kuroki K, Schommer SK, et al. Pathology in practice. Fran- 11. Rhyan JC, Gahagan T, Fales WH. Tularemia in a cat. J Vet Diagn
cisella tularensis. J Am Vet Med Assoc. 2011;238:1271-1273. Invest. 1990;2:239-241.
Weinberg AN, Branda JA. Case records of the Massachusetts General 12. Spagnoli ST, Kuroki K, Schommer SK, et al. Pathology in practice.
Hospital. Case 31-2010. A 29-year-old woman with fever after a cat Francisella tularensis. J Am Vet Med Assoc. 2011;238:1271-1273.
bite. N Engl J Med. 2010;363:1560-1568. 13. Gliatto JM, Rae JF, McDonough PL, et al. Feline tularemia on Nan-
tucket Island. Massachusetts. J Vet Diagn Invest. 1994;6:102-105.
REFERENCES 14. Inzana TJ, Glindemann GE, Snider G, et al. Characterization of a
1. McCoy GW, Chapin CW. Bacterium tularense, the cause of a plague- wild-type strain of Francisella tularensis isolated from a cat. J Vet
like disease in rodents. Public Health Bulletin. 1912;53:17-23. Diagn Invest. 2004;16:374-381.
2. Petersen JM, Molins CR. Subpopulations of Francisella tularensis 15. Valentine BA, DeBey BM, Sonn RJ, et  al. Localized cutaneous
ssp. tularensis and holarctica: identification and associated epide- infection with Francisella tularensis resembling ulceroglandular
miology. Future Microbiol. 2010;5:649-661. tularemia in a cat. J Vet Diagn Invest. 2004;16:83-85.
3. Kugeler KJ, Mead PS, Janusz AM, et  al. Molecular Epidemiol- 16. Magnarelli L, Levy S, Koski R. Detection of antibodies to Fran-
ogy of Francisella tularensis in the United States. Clin Infect Dis. cisella tularensis in cats. Res Vet Sci. 2007;82:22-26.
2009;48:863-870. 17. Lundstrom JO, Andersson AC, Backman S, et  al. Transstadial
4. Whipp MJ, Davis JM, Lum G, et al. Characterization of a novicida- transmission of Francisella tularensis holarctica in mosquitoes.
like subspecies of Francisella tularensis isolated in Australia. J Med Sweden. Emerg Infect Dis. 2011;17:794-799.
Microbiol. 2003;52:839-842. 18. Berrada ZL, Telford Iii SR. Survival of Francisella tularensis Type
5. Mahy S, Chavanet P, Piroth L, et  al. Emergence of tularemia A in brackish-water. Arch Microbiol. 2011;193:223-226.
in France: paradigm of the Burgundy region. Int J Infect Dis. 19. Santic M, Ozanic M, Semic V, et al. Intra-vacuolar proliferation of
2011;15:e882-e883. F. novicida within H. vermiformis. Front Microbiol. 2011;2:78.
6. Ellis J, Oyston PC, Green M, et al. Tularemia. Clin Microbiol Rev. 20. Ancuta P, Pedron T, Girard R, et  al. Inability of the Fran-
2002;15:631-646. cisella tularensis lipopolysaccharide to mimic or to antagonize
7. Baldwin CJ, Panciera RJ, Morton RJ, et al. Acute tularemia in three the induction of cell activation by endotoxins. Infect Immun.
domestic cats. J Am Vet Med Assoc. 1991;199:1602-1605. 1996;64:2041-2046.
8. Meinkoth KR, Morton RJ, Meinkoth JH. Naturally occurring tula- 21. Gunn JS, Ernst RK. The structure and function of Francisella lipo-
remia in a dog. J Am Vet Med Assoc. 2004;225:538:545-547. polysaccharide. Ann N Y Acad Sci. 2007;1105:202-218.
CHAPTER 56  Tularemia 545

22. Dai S, Mohapatra NP, Schlesinger LS, et al. The acid phosphatase 30. Seibold E, Bogumil R, Vorderwulbecke S, et al. Optimized appli-
AcpA is secreted in vitro and in macrophages by Francisella spp. cation of surface-enhanced laser desorption/ionization time-
Infect Immun. 2012;80:1088-1097. of-flight MS to differentiate Francisella tularensis at the level of
23. Barry EM, Cole LE, Santiago AE. Vaccines against tularemia. Hum subspecies and individual strains. FEMS Immunol Med Microbiol.
Vaccin. 2009;5:832-838. 2007;49:364-373.
24. Gustafson BW, DeBowes LJ. Tularemia in a dog. J Am Anim Hosp 31. Molins CR, Carlson JK, Coombs J, et  al. Identification of Fran-
Assoc. 1996;32:339-341. cisella tularensis subsp. tularensis A1 and A2 infections by real-
25. Weinberg AN, Branda JA. Case records of the Massachusetts Gen- time polymerase chain reaction. Diagn Microbiol Infect Dis.
eral Hospital. Case 31-2010. A 29-year-old woman with fever after 2009;64:6-12.
a cat bite. N Engl J Med. 2010;363:1560-1568. 32. Steward J, Piercy T, Lever MS, et  al. Treatment of murine
26. Penn RL. Francisella tularensis (tularemia). In: Mandell GL, Ben- pneumonic Francisella tularensis infection with gatifloxa-
nett JE, Dolin R, eds. Principles and Practice of Infectious Dis- cin, moxifloxacin or ciprofloxacin. Int J Antimicrob Agents.
eases. 7th ed. Philadelphia, PA: Churchill Livingstone, Elsevier; 2006;27:439-443.
2010:2927-2937. 33. Urich SK, Petersen JM. In  vitro susceptibility of isolates of Fran-
27. Capellan J, Fong IW. Tularemia from a cat bite: case report cisella tularensis types A and B from North America. Antimicrob Agents
and review of feline-associated tularemia. Clin Infect Dis. Chemother. 2008;52:2276-2278.
1993;16:472-475. 34. Conlan JW, Oyston PC. Vaccines against Francisella tularensis.
28. Splettstoesser WD, Seibold E, Zeman E, et al. Rapid differentiation Ann N Y Acad Sci. 2007;1105:325-350.
of Francisella species and subspecies by fluorescent in situ hybrid- 35. Rumble CT. Pneumonic tularemia following the shearing of a dog.
ization targeting the 23S rRNA. BMC Microbiol. 2010;10:72. J Med Assoc Ga. 1972;61:355.
29. Grunow R, Splettstoesser W, McDonald S, et  al. Detection of 36. Siret V, Barataud D, Prat M, et al. An outbreak of airborne tularae-
Francisella tularensis in biological specimens using a capture mia in France, August 2004. Euro Surveill. 2006;11:58-60.
enzyme-linked immunosorbent assay, an immunochromato- 37. Teutsch SM, Martone WJ, Brink EW, et al. Pneumonic tularemia
graphic handheld assay, and a PCR. Clin Diagn Lab Immunol. on Martha’s Vineyard. N Engl J Med. 1979;301:826-828.
2000;7:86-90.
CHAPTER 57

Bite and Scratch Wound Infections


Jane E. Sykes

Dogs
Overview of Bacterial Bite Wound Infections Bite wounds in dogs most often result from aggressive dog-dog
Causes: A variety of aerobic and anaerobic bacteria that include interactions. The dog breeds affected depend on regional variation
mycoplasmas and mycobacterial species. Pasteurella spp. in dog breed popularity. Younger adult, male dogs and especially
are commonly isolated. intact male dogs appear to be predisposed, but studies that compare
Geographic Distribution: Worldwide affected dogs to a control (or “background”) population have not
been reported.1-3 Bite wounds in dogs can also result from the bites
Major Clinical Signs: Fever, lethargy, inappetence, hyperes-
of other domestic and wild animal species, but the epidemiology
thesia, puncture wounds, cutaneous swellings, lacera-
of infections that result from these wounds is not well described.
tions, tissue avulsions, and signs that relate to damage
Approximately 20% of dog-dog bite wounds become
and/or infection of other structures (such as the thorax,
infected, as opposed to contaminated. The likelihood that a
abdomen, and brain and spinal cord)
wound becomes infected increases with the time lag between the
Differential Diagnoses: Other traumatic injuries, cutaneous neo- aggressive interaction and when veterinary attention is sought.2,3
plasia, sterile nodular panniculitis (dogs), plague, tularemia Infection develops 8 to 24 hours after the bite occurs and is less
Human Health Significance: Cat and dog bite wound infec- likely to be develop if the wound is limited to the dermis.3 The
tions are common in humans and may be life threatening distribution of bacterial species involved varies from one study
in the immunocompromised. They also can be associated to another and may be influenced by whether infected or only
with transmission of potentially serious bloodborne bac- contaminated wounds were evaluated. The most common bac-
terial diseases such as bartonellosis and tularemia. terial species isolated from dog-dog bite wounds are Staphylo-
coccus pseudintermedius, Enterococcus spp., Pasteurella spp.,
streptococci, and Escherichia coli.1-3 Staphylococcus aureus and
Capnocytophaga canimorsus, which are important pathogens
Etiology and Epidemiology in humans that are bitten by dogs, are rarely isolated from bit-
ten dogs,2,3 although the prevalence of C. canimorsus is proba-
Bite wounds are one of the most common reasons that dogs bly underestimated because it is very difficult to grow in culture.
or cats are brought to veterinary hospitals for care. Despite In the author’s hospital, Pasteurella spp. accounted for 17% of
this, there is surprisingly little information available on their 41 bacterial species cultured from 33 dog bite wounds, followed
epidemiology in comparison to the plethora of publications on by a variety of staphylococci (some methicillin-­resistant) (15%),
animal bite wounds in people (see Public Health Aspects). Bite E. coli (12%), Enterococcus spp. (7%), and Bacteroides spp.
wounds often result in bacterial infection, but can also result (7%). Other organisms were Peptostreptococcus spp., Clostrid-
in systemic viral infection (especially in the case of bitten cats), ium spp., and Pseudomonas aeruginosa (each 5%); and Strepto-
and less often, fungal or bloodborne protozoal infections such coccus viridans, Actinomyces spp., Acinetobacter spp., Serratia
as babesiosis (Table 57-1). marcescens, Klebsiella pneumoniae, Myroides spp., Prevotella
Bacterial pathogens that cause bite wound infections are spp., Fusobacterium spp., Aeromonas spp., Mycobacterium
usually members of the normal oral cavity flora of the biting smegmatis, and Mycoplasma spp. (one isolate each or 2%).
animal species, but organisms in the environment can also Obligate anaerobes were isolated from 5 dogs (15%), and in all
contaminate bite or scratch wounds (e.g., Clostridium tet- 5 of these dogs, multiple bacterial species were isolated.
ani, Mycobacterium spp., or Nocardia spp.). Bite wounds are
instantly contaminated with millions of bacterial species from Cats
the oral cavity, but only a small proportion of these species pos- Most bite wounds in cats result from aggressive interactions with
sess virulence properties that allow them to proliferate within other cats, but nonfatal dog bite wounds can also occur. Occasion-
the wound and cause disease. The degree to which proliferation ally, cats are bitten by a variety of other small wildlife species that
occurs also depends on host factors such as underlying immu- vary geographically based on the local fauna present. In contrast
nocompromise. The bacterial species that cause bite wound to dogs, which often crush and tear tissues with their teeth, cats
infections in dogs and cats differ from those that cause dog deliver deep puncture wounds that create an environment where
or cat bite wound infections in human patients, so data from obligate and facultative anaerobes flourish. Thus, anaerobic bac-
human studies cannot be extrapolated to dog or cat bite wound terial infections are more prevalent in cat bite abscesses than in
infections. dog bite wounds, and accordingly, polymicrobial infections are

546
CHAPTER 57  Bite and Scratch Wound Infections 547

TABLE 57-1
Pathogens That Cause Local or Systemic Infections in Dogs and Cats as a Consequence of Bite or Scratch Wounds from the Same or
Another Animal Species
Organism Class Dogs Cats
Viruses Rabies Rabies
FeLV
FIV
Mycoplasmas Hemotropic mycoplasmas? Nonhemotropic Mycoplasma spp.
Hemotropic mycoplasmas?
Gram-negative Pasteurella spp. Pasteurella spp.
aerobes* Escherichia coli Escherichia coli
Pseudomonas aeruginosa
Acinetobacter spp.
Proteus spp.
Enterobacter spp.
Serratia marcescens
Aeromonas spp.
Capnocytophaga canimorsus
Gram-positive Staphylococcus spp., especially S. pseudintermedius Streptococcus spp.
aerobes* Streptococcus spp. Enterococcus spp.
Enterococcus spp. Actinomyces spp.
Actinomyces spp. Nocardia spp.
Corynebacterium spp. Lactobacillus spp.
Corynebacterium spp.
Anaerobes Bacteroides spp. Bacteroides spp.
Clostridium spp. Prevotella spp.
Porphyromonas spp. Porphyromonas spp.
Fusobacterium spp. Fusobacterium spp.
Peptostreptococcus spp. Clostridium spp.
Propionibacterium spp. Propionibacterium spp.
Prevotella spp.
Eubacterium spp.
Mycobacteria Tuberculous mycobacteria (e.g., M. bovis in the Tuberculous mycobacteria (e.g., M. microti in the
United Kingdom) United Kingdom)
Rapidly growing mycobacteria Rapidly growing mycobacteria (e.g., M. fortuitum)
Lepromatous mycobacteria (e.g., M. lepraemurium)
Fungi Sporothrix schenckii Sporothrix schenckii
Opportunistic molds (e.g., Paecilomyces spp.), dematia-
ceous molds
Protozoa Babesia gibsoni None known
Babesia conradae?

*The term ‘aerobe’ refers to facultative anaerobes and obligate aerobes.

present more often in closed cat bite abscesses than in dog bite Clinical Features
wounds. In a study of 36 closed cat bite abscesses, 168 bacte-
rial strains were isolated, of which 72% were obligate anaerobes Dogs
and 28% were facultative anaerobes.4 The most prevalent anaer- Among dogs, dog bite wounds consist of abrasions, lacerations,
obes isolated from cat bite abscesses include Porphyromonas, avulsions (i.e., skin flaps), crushing injuries, and deep puncture
Bacteroides, Prevotella, Peptostreptococcus, and Fusobacterium. wounds (Figure 57-1). Abscesses can also develop. Dog bite
Pasteurella multocida, a commensal of the oral cavity of virtu- wounds may also penetrate body cavities and cause pneumotho-
ally all cats, is the most common facultative anaerobe present.4-6 rax or damage the esophagus, vertebral column, or gastrointesti-
Porphyromonas spp. appear to be particularly prevalent; in one nal tract. Pyothorax or bacterial peritonitis can also occur. Most
study of 15 abscesses in Australian cats, they accounted for 92% dogs have between 1 and 5 wounds. Rarely, more than 10 wounds
to 99% of all the facultative and obligate anaerobes present.7 are present.1-3 The majority of bite wounds occur cranial to the
Less often Actinomyces, β-hemolytic streptococci, lactobacilli, diaphragm, especially on the head and neck (Figure 57-2). The
and Propionibacterium spp. have been isolated. location of the bite wounds also depends on the size of the bitten
548 SECTION 2  Bacterial Diseases

A B

C
FIGURE 57-1  Dog bite wounds. A, Multiple puncture wounds, abrasions and lacerations to the abdomen of a 10-year-old female spayed mixed breed dog. A portion of the jejunum
had perforated and had herniated into the subcutaneous tissue. The dog developed cardiac arrest after 2 days of aggressive treatment that included surgery and was euthanized. B, Five-
year-old male neutered Pomeranian dog with a bite wound to the lateral cervical region from a pit-bull terrier dog that resulted in tissue avulsion. C, Inguinal region of a 6-year-old intact
male German shepherd dog with severe bite wounds and tissue avulsion that resulted in extensive exposure of muscle and bone. The dog had been attacked by four other dogs. The dog
survived with aggressive medical treatment and surgery that included amputation of the right pelvic limb. (Courtesy of the University of California, Davis Veterinary Emergency and Critical
Care Service.)

FIGURE 57-2  Common anatomic distribution of dog-to-dog bite and fight wounds.
CHAPTER 57  Bite and Scratch Wound Infections 549

dog; large-breed dogs are more likely to have wounds on the neck on the bridge of the nose, pinna, and inguinal region. Cat bite
and face, but small-breed dogs often have wounds on their dor- abscesses are characterized by the presence of firm or fluctu-
sum. Injury to underlying tissue is frequently dramatically more ant subcutaneous swellings or masses, with or without fever,
severe than is apparent on the surface. Classification systems have lethargy, inappetence, and hyperesthesia. Some cats are brought
been used to describe the severity of dog-dog bite wounds based to the veterinarian solely because of lethargy, and a careful
on the type of injury (laceration vs. puncture wound) and the physical examination is required before an abscess or celluli-
presence or absence of dead space or abscessation.1-3 Abscessa- tis is detected. One or more scabs may be found on top of the
tion is rarely associated with dog bite wounds as compared with abscess, or the overlying skin may lack hair and have a gray,
cat bite wounds.3 The presence of pus, fever, erythema, subcuta- necrotic appearance. If the abscess ruptures, cream-colored or
neous emphysema, and/or a foul odor suggests that infection has red-brown purulent material may be identified on the haircoat,
occurred. Uncommonly, hematogenous spread of bacteria leads sometimes in association with a foul odor (Figure 57-4). Alter-
to signs of severe sepsis or septic shock (see Chapter 86). natively, the haircoat may be matted over the site. Lameness
may be apparent if the limbs are involved, and especially if there
Cats is extension to the bone or a joint. Involvement of the thoracic
In cats, cat bite wounds may be difficult to identify on the sur- cavity may be associated with fever, lethargy, tachypnea, and
face, but infection can extend to penetrate bone, joints, ten- decreased lung sounds due to pyothorax (see Chapter 87). Bite
don and muscle.5 Cat bite wounds are most often located on wounds to the calvarium can result in neurologic signs such
the forelimbs, lateral aspect of the face, and near the base of as circling, disorientation, head pressing, mental obtundation,
the tail (Figure 57-3).8 In contrast, scratches are often found delayed or absent placing reactions, tetraparesis, anisocoria,

FIGURE 57-3  Common anatomic distribution of cat-to-cat bite and fight wounds (“wound cat”). (Adapted from Malik R, Norris J, White J, et al. “Wound cat.” J Feline Med Surg
2006;8:135-140.)

A B
FIGURE 57-4  A, Six-year-old female spayed domestic shorthair cat with a cat bite abscess adjacent to the right eye that was associated with marked chemosis and periocular swelling.
B, Purulent material discharged from the abscess when it was drained surgically. (Images courtesy University of California, Davis Veterinary Ophthalmology service.)
550 SECTION 2  Bacterial Diseases

absent menace responses, and seizures.9 Penetration of the Cats with dog bite wounds often have life-threatening inju-
caudal vertebral column and spinal cord can lead to vertebral ries. Fractures, severe hemorrhage, and hypovolemic shock
fractures, osteomyelitis, bacterial meningitis, and signs of pelvic can dominate the clinical picture. Penetrating injury of cervical
limb paralysis (Figure 57-5). As in dogs, progression to severe structures, the thorax, abdominal organs, brain, or spinal cord
sepsis or septic shock can occur, but most cat bite abscesses can occur (Figure 57-6). Death most often results from trauma
rupture and resolve spontaneously. rather than infection, but occasionally deep-seated infections
or bacteremia develops if the cat survives the initial traumatic
episode.

Diagnosis
A diagnosis of bite wound infections is based on history of a fight
or bite and physical examination findings that suggest that infec-
tion has developed. Cytologic examination of a wound may also
assist in diagnosis of infection. The veterinarian should record
whether the wound was provoked or unprovoked and note time
and location that the wound occurred and the biting animal
species involved. Depending on the extent and location of bite
wounds, radiographs of the affected region should be consid-
ered to assess for underlying damage to bone or body cavities.
All cats with cat fight wounds should be tested for FeLV and
FIV infection, and the test should be repeated 2 months later
because transmission may have occurred at the time of the fight
wound (see Chapters 21 and 22).
FIGURE 57-5  Histopathology of the vertebral column of a young adult, male domes- Laboratory Abnormalities
tic shorthair that was found with a bite wound to the base of the tail that drained purulent
material and pelvic limb paralysis with absent deep pain sensation. Multiple fractures Laboratory abnormalities in cats or dogs with wound infec-
of caudal vertebrae 4 and 5 were present in association with suppurative osteomyeli- tions are variable and depend on the degree of tissue trauma,
tis (arrowheads), cellulitis, ascending meningitis, and masses of intralesional bacteria the underlying organs involved (if any), and the severity and
(arrows). Multiple anaerobes were cultured from the lesion. H&E stain. type of infection. The CBC may show nonregenerative or regen-
erative anemia, neutrophilia with bandemia, toxic neutrophils,

B
FIGURE 57-6  A, Lateral radiograph of the lumbar spine of a 3-year-old female Siamese cat with a dog bite wound to the lumbar spine. The cat was unable to stand and had absent
placing reactions in the pelvic limbs. Patellar reflexes were also absent. Two osseous densities are superimposed at the level of the dorsal spinal canal at the level of the fourth lumbar ver-
tebra (L4) (arrow). The dorsoventral view showed deviation of the spinous process of L4 (not shown). CSF analysis revealed mild suppurative inflammation. B, Myelogram. A spinal needle
is inserted at L5-6 for injection of contrast agent. Spinal cord widening with associated thinning of the contrast column circumferentially is identified over the body of L4. Dorsal deviation
and splitting of the ventral contrast column is present over L2-3 as a result of disc protrusion in this region. A decompressive hemilaminectomy over L4 to L6 was performed and a bone
fragment was removed; the cat ultimately recovered.
CHAPTER 57  Bite and Scratch Wound Infections 551

monocytosis, and lymphopenia or lymphocytosis. Animals with with a needle and syringe can avoid contamination by sur-
severe sepsis or septic shock may be thrombocytopenic. Findings face bacteria. Because a positive culture from a wound does
on the chemistry panel include hypoalbuminemia and evidence not imply infection, results must be interpreted in light of the
of muscle damage (increased activities of CK, AST, and some- physical examination and laboratory findings. Susceptibility test
times mild increases in serum ALT activity). Increased muscle results for isolated bacteria can then be used to guide antimi-
enzyme activities may be a clue to an underlying cat bite wound crobial drug treatment. Blood cultures should be considered for
in cats with fever of unknown origin. Prolongations of the PT dogs with evidence of severe sepsis or septic shock.
and/or APTT may be present in animals with septic shock and
disseminated intravascular coagulation. Isolation and Identification: Cats
In general, culture of cat bite abscesses in cats is not routinely
Diagnostic Imaging performed because they almost always respond to drainage with
Plain Radiography or without systemic antibacterial drug treatment. Cytologic
Radiographs of the thorax in dogs or cats with bite wounds examination and culture are indicated for cat bite abscesses/
may show fractured ribs, pneumothorax, evidence of pulmo- cellulitis if clinical signs of infection are persistent or recur-
nary contusions, mediastinal emphysema, diaphragmatic her- rent in the face of treatment, or if severe infections that involve
niation, or pleural effusion due to hemothorax or pyothorax. underlying bones, joints, or body cavities occur. Culture is also
Abdominal radiographs may show a lack of serosal detail due recommended for draining skin lesions in the inguinal region,
to hemoabdomen or bacterial peritonitis. Radiographs of the which may result from infection with rapidly growing mycobac-
axial or appendicular skeleton may reveal fractures or tooth for- teria (see Chapter 44). Blood cultures are indicated for cats with
eign bodies. In more chronic bite wound infections, evidence of signs of severe sepsis or septic shock.
osteomyelitis or septic arthritis may be present, with soft tissue
swelling and bony lysis (see also Chapter 85). Treatment and Prognosis
Sonographic Findings Surgical Management: Dogs
Ultrasound of soft tissues can be useful to assess the extent of Dogs with dog bite wounds should be muzzled and the wound(s)
bite wound infections. Abdominal ultrasound is indicated for covered with a clean, dry bandage immediately after the injury
animals with bite wounds to the abdomen to assess for evidence occurs to prevent further contamination of the wound until
of peritonitis or trauma to abdominal organs. Findings in dogs the animal’s condition can be stabilized if necessary. If possi-
or cats with peritonitis include a hyperechoic mesentery with ble, the dog should then be sedated or anesthetized and a wide
an irregular outline and free peritoneal fluid, which is typically area of skin around the wound clipped in order to determine
echogenic (see Chapter 88). the extent of injury and prepare for surgery. Frequently, this
leads to detection of additional full-thickness puncture wounds
Advanced Imaging or lacerations. The skin around the wound should be scrubbed
Findings on MRI in cats with brain abscesses secondary to with chlorhexidine or povidone-iodine solution. Full-thickness
cat bite wounds include space-occupying lesions with well- punctures and lacerations should then be explored, debris and
defined margins that are hyperintense on T2-weighted images, devitalized tissue removed, and specimens collected for culture
and hypointense T1-weighted images.9 Marked ring enhance- if infection is suspected. If the abdominal or thoracic cavity has
ment is usually present. Evidence of brain herniation may be been penetrated, surgical exploration of these cavities is typically
detected.9 required to identify and treat damaged viscera. Wounds should
be lavaged extensively with copious quantities of sterile saline
Cytologic Examination or lactated Ringer’s solution under moderate pressure (such as
Cytologic examination of swab or fluid specimens collected using a 60-mL syringe attached to an 18-gauge needle). The use
from infected wounds typically reveals large numbers of degen- of antibiotics or antiseptics to lavage the wound is not gener-
erate neutrophils with intracellular bacteria, which may include ally recommended, as they can be toxic to tissue and provide
cocci, rods, or long, filamentous organisms (which are usually minimal benefit for treatment of established wound infections.
anaerobes). If used, 0.05% chlorhexidine diacetate (1 part 2% chlorhexi-
dine to 39 parts water) is preferred because it has antibacterial
Microbiologic Tests activity in the presence of organic debris, residual antibacterial
Isolation and Identification: Dogs activity, and minimal systemic absorption and toxicity.
Aerobic bacterial culture and susceptibility and anaerobic bac- In general, infected wounds that are limited to the subcu-
terial culture are indicated whenever possible from dogs that taneous tissues and muscle are subsequently managed as open
are suspected to have infected bite wounds. Culture lacks sensi- wounds in order to optimize drainage and debridement. Closure
tivity for detection of anaerobes, and anaerobic bacterial infec- of infected wounds can lead to dehiscence and persistence of
tions respond predictably to treatment, so anaerobic culture infection. Whenever possible, the wound is covered with ban-
could be considered optional if client finances are limited (see dages that absorb fluid, reduce dead space, prevent movement
Chapter 37). and contamination of the wound, and assist in debridement.
The skin around the wound should be clipped and cleaned Hydrophilic bandages are often used for the first 3 to 5 days.
with chlorhexidine solution, and an aspirate, swab, or devital- Wound closure may be possible for noninfected wounds that
ized tissue specimen should be collected from as deep within the are in the “golden period” (first 6 to 8 hours after injury). The
wound as possible while the animal is sedated or anesthetized. reader is referred to small animal surgical texts for additional
When pockets of purulent material extend subcutaneously away information on surgical management of bite wound infections
from an open wound, collection of aspirates percutaneously and bandaging.10
552 SECTION 2  Bacterial Diseases

TABLE 57-2
Suitable Antimicrobial Drugs for the Treatment of Bite Wound Infections in Dogs and Cats*
Drug Species Dose (mg/kg) Interval (hours) Route
Amoxicillin-clavulanic acid D, C 12.5-20 8-12 PO
Ampicillin-sulbactam D, C 10-20† 8 IV, IM
Pradofloxacin‡ D 3 24 PO
C 5-7.5 24 PO
Enrofloxacin D 5-20 24 IV, IM, PO
Marbofloxacin D 2.75-5.5 24 PO
Gentamicin D 9-14 24 IV, IM
Amikacin D 15-30 24 IV, IM
Metronidazole D, C 10 8 PO, IV

C, Cat; D, dog.
*Doses of antimicrobial drugs that have activity primarily against gram-negative aerobes are listed for dogs, because isolation of gram-negative aerobes
from cat bite abscesses is very rare.
†Dose according to ampicillin component.
‡The pradofloxacin dose for dogs is for the tablet formulation available in Europe. The dose for cats is for the oral suspension. The use of pradofloxa-

cin in dogs may be associated with myelosuppression and is off-label in some countries.

Surgical Management: Cats and an aminoglycoside, or a combination of ampicillin, met-


As for open dog bite wounds, a wide area of skin around cat bite ronidazole, and an aminoglycoside (Table 57-2). Where
abscesses should be clipped and scrubbed. The abscess should methicillin-resistant S. pseudintermedius is not prevalent, a fluo-
then be lanced and specimens collected for culture if desired. roquinolone could be substituted for the aminoglycoside. Sub-
The abscess cavity should then be lavaged, necrotic tissue sequent treatment should be based on the results of culture and
debrided, and a placement of a drain should be considered to susceptibility. Amoxicillin-clavulanic acid is the drug of choice
optimize drainage and prevent early wound closure. Whenever for treatment of cat bite abscesses once drainage has been estab-
possible, the wound and drain should then be covered with a lished (although many cat bite abscesses will likely heal with
bandage. The drain is removed in 3 to 5 days. For cats with pyo- drainage alone). Because Pasteurella spp. are not susceptible to
thorax, placement of chest tubes with or without surgical explo- clindamycin or first-generation cephalosporins, monotherapy
ration may be required (see Chapter 87). Cats with abscesses with these drugs should be avoided. Pradofloxacin is also a suit-
that involve the central nervous system have the potential to able choice, because it has activity against anaerobes as well as
recover with craniectomy, drainage of purulent material within a variety of aerobic bacterial species.
the abscess, removal of any tooth fragments present, and clo- Antimicrobial drug treatment of infected bite wounds that
sure of subcutaneous tissue and the skin.9 are localized to the skin, subcutaneous tissue, and muscle
should continue for 3 to 5 days after signs of infection are no
Antimicrobial Treatment longer evident. The reader is referred to Chapters 85, 87, and
Contaminated Wounds (First 8 Hours after Injury) 88 for information on the management of osteomyelitis, septic
The need for antimicrobial treatment in contaminated, nonin- arthritis, pyothorax, and peritonitis.
fected wounds is controversial, because there is no evidence that
it prevents infection of the wound and it may select for infec- Rabies Prophylaxis
tion with bacteria that are resistant to the antimicrobial drug In regions where rabies occurs, bite wounds may need to be
used. Proper wound cleaning and debridement are likely to have reported to local public health authorities. If the biting animal has
the greatest impact on whether infection subsequently develops. an unknown vaccination history, or the bite was from a wild ani-
Antimicrobial treatment could be considered for deep punctures mal that was unavailable for testing, a rabies booster should be
or large, dirty wounds that require extensive debridement. If administered and the animal monitored for 45 days. If the bitten
deemed necessary, the drug of choice for contaminated wounds animal is unvaccinated or has a lapsed vaccination status, addi-
is amoxicillin–clavulanic acid, because it has efficacy against tional quarantine or euthanasia may be required (see Chapter 13).
most oral cavity commensals of the dog and cat mouth such as
Pasteurella spp. and a variety of anaerobes, including those that Prognosis
produce β-lactamase enzymes. The prognosis for bite wound infections depends on the severity
and extent of injury, the immune status of the host, and the organ-
Infected Wounds isms involved. Recurrent infections may result from underlying
For severely infected dog bite wounds, initial treatment should osteomyelitis, foreign bodies, immunodeficiency, or the presence
consist of parenteral antimicrobial drugs that have activity of atypical organisms such as Mycobacterium spp., Nocardia
against gram-negative and gram-positive aerobic and anaerobic spp., or Mycoplasma spp. (which do not respond to β-lactam
bacteria, which includes methicillin-resistant staphylococci (in drugs). Failure to respond to appropriate treatment should
regions where methicillin-resistant S. pseudintermedius is prev- prompt a thorough physical examination; testing for retroviruses
alent). Appropriate combinations include ampicillin-­sulbactam in cats; imaging of the affected area; and culture for aerobic and
CHAPTER 57  Bite and Scratch Wound Infections 553

anaerobic bacteria and mycoplasmas. If the clinical picture is sug- more rapidly than dog bite wounds.14 The upper extremities
gestive, culture for mycobacteria may also be indicated. are often affected. As in dog bite wounds, Pasteurella spp. are
the most common organisms isolated and are present in 75% of
Prevention wounds.14,19 The predominant species are P. multocida subsp.
multocida and P. multocida subsp. septica. Streptococci (includ-
Prevention of bite wounds involves education of pet owners in ing S. pyogenes) and staphylococci are also often present, but S.
regard to housing cats indoors and use of secure collars and leashes aureus is isolated less frequently from cat bite wounds than dog
for walking dogs. Intact male dogs should be neutered if they are bite wounds. Neisseria and Moraxella are also often isolated
not to be used for breeding. Proper handling and socialization of from cat bite wounds (see Table 57-3). The prevalence of infec-
puppies as well as behavior training should be encouraged. Pet tion with anaerobes in cat bite wounds is similar to that in dog
owners should be instructed to avoid or closely supervise inter- bite wounds.14
actions between their dogs and unfamiliar dogs that are breeds
of known aggressive tendency, such as American pit bull terriers, Clinical Manifestations
Rottweilers, German shepherds, blue heelers, chow chows, and Infection of bite wounds in humans most often results in localized
Jack Russell terriers. Special caution is warranted for small-breed cellulitis, pain, purulent discharge, a black to gray appearance,
dogs, which may provoke larger breed dogs and are more likely and, less often, fever and local lymphadenopathy. Extension to
to develop life-threatening injuries. Dogs that are unfamiliar with underlying bones or joints can lead to osteomyelitis or septic
one another or dogs with a history of fighting with one another arthritis, which is especially likely to occur after penetrating cat
should never be left together without proper supervision. bite wounds to the hand (Figure 57-7). Rarely, necrotizing fas-
ciitis, bacteremia, meningitis, endocarditis, peritonitis, or brain
Public Health Aspects abscesses develop.19,28
Most of the serious consequences of dog or cat bite wound
Epidemiology of Dog and Cat Bite Wounds infections occur in immunocompromised humans and involve
In 2012, the American Pet Products Association estimated that P. multocida or Capnocytophaga canimorsus. Severe disease
there are 78.2 million owned dogs and 86.4 million owned cats manifestations have also occurred after exposure of wounds
in the United States, and that 39% of households own at least to dog or cat saliva through licking, or after invasive medical
one dog and 33% own at least one cat.11 In the United Kingdom, supplies (such as dialysis tubing) were chewed or licked by
around 22% of households own a dog and 18% own a cat.12 Ger- a pet.29 In a few cases, patients have had a history of con-
man shepherds, Rottweilers, and Jack Russell terriers are among tact with animals but no known bite or scratch wound.30-32
the top 10 dog breeds owned. Dog and cat bite wounds account Life-threatening infections with P. multocida tend to occur
for approximately 1% of emergency department visits by humans in infants, pregnant women with animal contact, patients
in the United States, and similar numbers are reported in Europe. on chronic glucocorticoid therapy, HIV-infected people,
Dog bites accounted for more than 70% and cat bites accounted organ-transplant recipients, and patients with chronic liver
for 13% of animal bite visits to emergency rooms in New York disease.28
City.13 The median age of bitten people in one study was 15 years, Capnocytophaga canimorsus is a capnophilic (carbon
and in another study it was 28 years.14,15 Many bites occur in chil- dioxide–loving), facultatively anaerobic, filamentous gram-
dren, especially boys aged 5 to 9 years.16 Children often have dog negative bacterial rod that is commonly found in the oral
bites to the face, neck, and head. Women and the elderly are at cavity of dogs and cats.33 Although rarely reported as a
risk for cat bite injuries. Patients who seek medical attention more cause of illness in dogs or cats, this organism can cause cel-
than 8 hours after injury usually have infected wounds. lulitis, gangrene, bacteremia, meningitis, and endocarditis in
humans.28,33 C. canimorsus bacteremia is rare, but humans
Dog Bite Infections who are immunocompromised, especially those with chronic
Between 3% and 20% of dog bite wounds in humans become alcoholism, cirrhosis, or a history of splenectomy, are at risk
infected.17,18 The infecting organisms generally reflect the flora for life-threatening C. canimorsus infections that progress rap-
of the canine oral cavity, but may also derive from the environ- idly, with a mortality of 31%. The virulence factors that con-
ment or the bitten person’s skin or oral cavity. Many infections tribute to these severe infections are not well understood. The
are polymicrobial, but the organism that most often contributes organism does not cause endotoxemia but appears to evade
to dog bite wound infections is Pasteurella spp., which is found recognition by the immune system and resist phagocytosis.28
in 50% of wounds (Table 57-3).14,19-27 The most common spe- In one series of humans with C. canimorsus sepsis, 56% of
cies is P. canis, but P. multocida (subspecies multocida, septica, cases followed a dog bite and 10% of cases followed expo-
and gallicida), P. stomatis, and P. dagmatis may also be present. sure of a wound to dog saliva by licking.34 Veterinarians have
Other common isolates from dog bite wounds include strepto- also developed corneal infections with C. canimorsus when
cocci (especially the α-streptococcus S. mitis and S. pyogenes; dog tooth fragments have struck them in the eye during den-
S. canis is rarely isolated), staphylococci, Corynebacterium tal extractions.35,36 Most affected humans are older than 40
spp., Neisseria spp., and anaerobes.19 Of the staphylococci, years of age. Clinical manifestations of C. canimorsus sepsis
S. aureus is isolated most often (20% of infections). Infections include fever, myalgia, and a petechial rash that involves the
with S. pseudintermedius occur rarely. Anaerobes are present in skin and mucous membranes, which may progress from pur-
approximately 70% of infections and are almost always present pura (purpura fulminans) to gangrene over a period of several
in combination with other anaerobes and aerobic bacteria. days. Because C. canimorsus is very difficult to grow, early
diagnosis and successful treatment require clinical suspicion
Cat Bite Infections and early treatment with effective antimicrobial drugs, such as
Although cat bite wounds are less traumatic to tissue, they β-lactam/β-lactamase inhibitor combinations, which are also
are more likely to become infected and progress to infection active against Pasteurella spp.
554 SECTION 2  Bacterial Diseases

TABLE 57-3
Pathogens That Can Infect Humans after a Bite from a Dog or Cat14,19-27
Dog Cat
Viruses Rabies Rabies
Aerobic Gram-positive bacteria Streptococcus (S. mitis)* Streptococcus (S. mitis)
Staphylococcus (S. aureus) Staphylococcus (S. epidermidis)
Corynebacterium (group G) Corynebacterium (C. aquaticum)
Enterococcus Enterococcus (E. durans)
Bacillus Bacillus (B. firmus)
Gemella morbillorum Actinomyces
Actinomyces viscosus Gemella morbillorum
Lactobacillus Erysipelothrix
Dermabacter hominis Rothia dentocariosa
Oerskovia Lactobacillus
Micrococcus lylae Rhodococcus spp.
Pediococcus Streptomyces spp.
Stomatococcus mucilaginosus
Aerobic Gram-negative bacteria Pasteurella (P. canis) Pasteurella (P. multocida)
Neisseria (N. weaveri) Neisseria (N. weaveri)
Moraxella Moraxella (M. catarrhalis)
Bergeyella zoohelcum Bergeyella zoohelcum
Capnocytophaga Capnocytophaga
Escherichia coli Acinetobacter
Brevibacterium Pseudomonas
Stenotrophomonas maltophilia Brevibacterium
Pseudomonas Gemella morbillorum
Klebsiella Actinobacillus
Citrobacter Alcaligenes
Flavobacterium Enterobacter
Eikenella corrodens Riemerella anatipestifer
Bartonella? Klebsiella oxytoca
Eikenella corrodens
Flavimonas oryzihabitans
Aeromonas hydrophila
Pantoea agglomerans
Bartonella
Francisella tularensis
Yersinia pestis
Anaerobic bacteria Fusobacterium (F. nucleatum) Fusobacterium (F. nucleatum)
Bacteroides (B. tectum) Bacteroides (B. tectum)
Prevotella (P. heparinolytica) Prevotella (P. heparinolytica)
Porphyromonas (P. macacae) Porphyromonas (P. gulae)
Propionibacterium (P. acnes) Propionibacterium (P. acnes)
Peptostreptococcus Peptostreptococcus
Tannerella forsythia Filifactor villosus
Eubacterium spp. Eubacterium spp.
Campylobacter spp. Clostridium sordellii
Veillonella spp.
Mycoplasmas Mycoplasma spp.
Mycobacteria Mycobacterium fortuitum Mycobacterium fortuitum
Mycobacterium kansasii
Fungi Blastomyces dermatitidis Sporothrix schenckii
Paecilomyces lilacinus

*For organisms that are present in >10% of cases, the most common species isolated is listed in parentheses.
CHAPTER 57  Bite and Scratch Wound Infections 555

Prevention
Veterinarians play an important role in educating the public
about prevention of cat and dog bite wounds. Provision of infor-
mation to clients who intend to purchase a new dog about dog
breeds, puppy socialization, dog behavior training, and how to
prevent and manage aggression and avoid bites or scratches (to
themselves and to others) is encouraged. Veterinarians can also
engage in community activities (especially in schools) where
young children can be educated about pet ownership and how
to avoid wounds and wound infections from their own or some-
one else’s dog or cat. Immunocompromised individuals should
be educated about the potential benefits and risks of pet owner-
ship and precautions that can be taken to minimize infections.
Finally, care should be taken in the veterinary practice situation
to avoid bites or scratches to veterinary staff. Veterinary staff
should be educated about the types of infections that can occur
in the workplace; likelihood of these infections; risk factors for
infection; appropriate preventative measures; and protocols to
follow should a bite or scratch wound occur (see General Ani-
mal Handling Precautions, Chapter 11). Routine hand-­washing
practices should be enforced, and licking by dogs or cats should
be discouraged. Face protection should be worn by veterinary
staff when dental procedures are performed. Bites and scratches
may need to be reported to public health authorities, and all
FIGURE 57-7  Cat bite wounds to the base of the thumb of a veterinarian with asso- staff should be educated about the need to ensure that tetanus
ciated cellulitis. Penetrating cat bite wounds of the hand have a tendency to extend to and rabies immunizations are not overdue.
underlying bones and joints.

CASE EXAMPLE Musculoskeletal: BCS 6/9, ambulatory.


Cardiovascular, Respiratory, Gastrointestinal, Genitourinary,
Lymph Nodes, and Neurologic: No clinically significant
Signalment: “Murphy”, a 5-year-old intact male boxer dog abnormalities were detected. Cranial nerve examination
from Vacaville in northern California was unremarkable. A full neurologic examination was not
History: Murphy was evaluated by the University of California, performed.
Davis, veterinary emergency service for dog bite injuries after Initial Treatment: Murphy was sedated with hydromorphone
a fight with another dog in the household. The owner had and dexmedetomidine. The left neck was clipped with a no.
returned home a few hours earlier to find Murphy bleeding 40 clipper blade from the midpoint of the dorsal aspect of
from the neck. The other dog, a 5-year-old male neutered the head to the ventral aspect of the left ear and the tho-
boxer mix, was found hiding and unable to stand with several rax. The skin was cleaned with 0.05% chlorhexidine solu-
“scratches” on his body. Murphy had a history of fights with tion, and all puncture sites were probed and flushed with
other dogs. Both dogs in the household were fully vaccinated sterile saline. The sites were found to be relatively superfi-
for canine distemper virus, parvovirus, adenovirus, and rabies. cial. Drain sites were created with a no. 11 scalpel blade for
There was no other pertinent medical history. three wounds, and a 3 × 0.75 inch Penrose drain was placed
Initial Physical Examination: across a wound site that had the greatest amount of dead
Body Weight: 46.2 kg. space and secured with a single interrupted suture. The dog
General: Quiet, alert, responsive and hydrated. T = 102.1°F was sent home with instructions for the owner to administer
(38.9°C), HR = 110, panting, pink and moist mucous amoxicillin-clavulanic acid (16 mg/kg PO q12h) and trama-
membranes, CRT = 1 s. dol (2.4 mg/kg PO q8-12h), to monitor the wound for devel-
Integument: The haircoat was full, thick, and dirty. There was opment of excessive discharge, bleeding, swelling, or red-
a 1-cm superficial laceration on the ventral aspect of the ness, and to keep the area clean with a washcloth soaked in
pinna, and approximately 10 puncture wounds on the left warm water. Instructions were provided to return in 3 days
ventral cervical region. A 2-cm superficial laceration was to have the wound reevaluated and the drain removed.
present at the cranial aspect of the dorsal midline, and a The owners returned 7 days later for drain removal. At that time,
5-cm superficial laceration was present on the left flank. No they reported that Murphy had been eating normally, but
ectoparasites were seen. had been drinking more than usual. His activity level had
Eyes, Ears, Nose, and Throat: Bilateral conjunctival hyperemia been normal, but they believed his neck was had become
and a mucoid ocular discharge were present. No other swollen and tender. They had been administering the medi-
significant abnormalities were noted. cations as directed.

Continued
556 SECTION 2  Bacterial Diseases

Physical Examination (Day 7): Isolate 3: Pseudomonas aeruginosa, resistant to ampicillin,


Body Weight: 46.2 kg. amoxicillin-clavulanic acid, all cephalosporins tested, chlor-
General: Quiet, alert, responsive, and hydrated but anxious. T = amphenicol, and doxycycline. Susceptibility to clindamycin
103.1°F (39.5°C), HR = 114, RR = 36, pink and moist mucous was not reported because this species is usually resistant.
membranes, CRT 1 s. Susceptible only to amikacin (≤4 µg/mL), gentamicin (2 µg/
Integument: There were marked crusts and scabs on the left mL), enrofloxacin (0.50 µg/mL), imipenem (≤1 µg/mL), ticar-
lateral and ventral aspect of the neck. The Penrose drain cillin (16 µg/mL), ticarcillin–clavulanic acid (16 µg/mL). The
was present with minimal amounts of discharge. Serous to MICs for trimethoprim-sulfamethoxazole and marbofloxacin
purulent discharge was draining from the ventral puncture were 2 µg/mL and ≤0.25 µg/mL, respectively.
wounds. There was moderate crusting of the dorsal aspect Anaerobic culture: Moderate numbers of mixed growth that
of the left pinna. included Bacteroides ureolyticus group (β-lactamase nega-
Other Systems: No change from the previous examination. tive), Peptostreptococcus anaerobius, and Bacteroides
Treatment (Day 7): The dog was again sedated and purulent spp./Prevotella spp. (β-lactamase negative).
material was collected for culture and susceptibility. The Diagnosis: Polymicrobial bite wound infection with anaerobes
eschars were gently removed with forceps and the skin and multiple drug-resistant aerobic gram-positive and
scrubbed with 0.05% chlorhexidine and water. Ultrasound gram-negative bacteria.
of the neck region revealed pockets of hypoechoic material Outcome: On receipt of the culture and susceptibility test
beneath the skin, and surgical debridement of the area results, the clindamycin was substituted with amoxicillin-
under general anesthesia was recommended. Subsequently clavulanic acid (16 mg/kg PO q12h). Treatment with
Murphy was anesthetized and the wound was explored. No enrofloxacin was continued and the owners were
abscesses were found. The devitalized tissue was excised, educated in regard to the public health significance of the
the wounds flushed vigorously, and two Penrose drains were infection and the need to wear gloves when handling the
placed. The neck was bandaged initially, but this interfered wound and for scrupulous hand-washing practices. The dog
with the dog’s ability to breathe comfortably, so the returned 4 days after surgery for drain removal. The owners
bandage was removed. The dog was hospitalized overnight reported that the drains had been producing considerable
and treated with hydromorphone (0.05 mg/kg, IV, q4h) and amounts of purulent material and that Murphy was doing
warm compresses to the neck (q6h for 5 minutes). He was well. He was afebrile and the wound was unchanged in
then sent home with instructions to administer tramadol as appearance. He was again sedated, the area was scrubbed,
previously prescribed, clindamycin (20 mg/kg PO q12h), and and crusts were removed. The drains were removed. Murphy
enrofloxacin (10 mg/kg PO q24h), and to warm compress recovered uneventfully and the sutures were removed 10
the neck every 4 to 6 hours for the next 3 days. They were days later at a local veterinary clinic. Five months later he
also instructed to return in 3 to 5 days for reevaluation and was seen again for multiple bite wounds to the thoracic
drain removal. limbs and right thorax.
Microbiologic Testing: Comments: In this dog, bite wound injuries were complicated
Direct Smear and Gram Stain: Rare numbers of gram-positive by infection with multiple gram-negative and gram-positive
cocci. drug-resistant aerobic bacteria as well as anaerobes. The
Aerobic and Anaerobic Culture and Susceptibility (Swab S. aureus and P. aeruginosa were resistant to amoxicillin-
from Wound): Isolate 1: Methicillin-resistant Staphylococcus clavulanic acid, which had been used to prevent infection
aureus, resistant to all β-lactams and clindamycin; when the dog was first seen. In contrast, the anaerobes and
susceptible to amikacin (≤4 µg/mL), chloramphenicol (8 the E. faecium were susceptible to amoxicillin-clavulanic
µg/mL), doxycycline (≤2 µg/mL), enrofloxacin (≤0.25 µg/ acid, but presumably were able to proliferate and persist
mL), gentamicin (≤1 µg/mL), marbofloxacin (≤0.25 µg/mL), in the face of antimicrobial drug treatment as a result of
rifampin (≤1 µg/mL), and trimethoprim-sulfamethoxazole the presence of the other drug-resistant bacteria, purulent
(≤0.50 µg/mL). material, and dead space. Bite wounds to the neck can be
Isolate 2: Enterococcus faecium (intrinsic resistance to low- a challenge to manage because of the mobility of this area,
level aminoglycosides, cephalosporins, clindamycin, fluo- which contributes to the maintenance of the dead space.
roquinolones, trimethoprim-sulfamethoxazole), resistant The use of a bandage can reduce dead space, avoid further
to chloramphenicol and rifampin. Intermediate suscepti- contamination, and reduce exposure to multidrug-resistant
bility to doxycycline (8 µg/mL), erythromycin (1 µg/mL), bacteria, but attempts to bandage the wound were not
ticarcillin (64 µg/mL), and ticarcillin-clavulanate (64 µg/ tolerated by this dog. Client education was an important part
mL). Susceptible to ampicillin (1 µg/mL), clavulanic acid– of management. The owners were instructed to separate the
amoxicillin (≤4 µg/mL), penicillin (4 µg/mL), and imipenem dogs when unattended to avoid further fights. Neutering
(≤1 µg/mL). may also have been of benefit.
CHAPTER 57  Bite and Scratch Wound Infections 557

SUGGESTED READINGS 16. Nonfatal dog bite-related injuries treated in hospital emergency
departments—United States, 2001. MMWR Morb Mortal Wkly
Gaastra W, Lipman LJ. Capnocytophaga canimorsus. Vet Microbiol. Rep. 2003;52:605-610.
2010;140:339-346. 17. Goldstein EJ. Bite wounds and infection. Clin Infect Dis.
Love DN, Malik R, Norris JM. Bacteriological warfare amongst cats: 1992;14:633-638.
what have we learned about cat bite infections? Vet Microbiol. 18. Underman AE. Bite wounds inflicted by dogs and cats. Vet Clin
2000;74:179-193. North Am Small Anim Pract. 1987;17:195-207.
Malik R, Norris J, White J, et  al. Wound cat. J Feline Med Surg. 19. Abrahamian FM, Goldstein EJ. Microbiology of animal bite wound
2006;8:135-140. infections. Clin Microbiol Rev. 2011;24:231-246.
Oehler RL, Velez AP, Mizrachi M, et  al. Bite-related and septic syn- 20. Zendri E, Martignoni G, Benecchi M, et al. Paecilomyces lilacinus
dromes caused by cats and dogs. Lancet Infect Dis. 2009;9:439-447. cutaneous infection associated with a dog bite. J Am Acad Derma-
tol. 2006;55:S63-S64.
REFERENCES 21. Ngan N, Morris A, de Chalain T. Mycobacterium fortuitum infec-
1. Griffin GM, Holt DE. Dog-bite wounds: bacteriology and treatment tion caused by a cat bite. N Z Med J. 2005;118:U1354.
outcome in 37 cases. J Am Anim Hosp Assoc. 2001;37:453-460. 22. Southern Jr PM. Tenosynovitis caused by Mycobacterium kansasii
2. Meyers B, Schoeman JP, Goddard A, et al. The bacteriology and associated with a dog bite. Am J Med Sci. 2004;327:258-261.
antimicrobial susceptibility of infected and non-infected dog bite 23. Ariel I, Haas H, Weinberg H, et  al. Mycobacterium fortuitum
wounds: fifty cases. Vet Microbiol. 2008;127:360-368. granulomatous synovitis caused by a dog bite. J Hand Surg Am.
3. Mouro S, Vilela CL, Niza MM. Clinical and bacteriological assess- 1983;8:342-343.
ment of dog-to-dog bite wounds. Vet Microbiol. 2010;144:127-132. 24. Gnann Jr JW, Bressler GS, Bodet 3rd CA, et al. Human blastomy-
4. Love DN, Jones RF, Bailey M, et al. Isolation and characterisation cosis after a dog bite. Ann Intern Med. 1983;98:48-49.
of bacteria from abscesses in the subcutis of cats. J Med Microbiol. 25. Thornton DJ, Tustin RC, Pienaar BJ, et  al. Cat bite transmis-
1979;12:207-212. sion of Yersinia pestis infection to man. J S Afr Vet Assoc.
5. Love DN, Malik R, Norris JM. Bacteriological warfare amongst 1975;46:165-169.
cats: what have we learned about cat bite infections? Vet Micro- 26. Yuen JC, Malotky MV. Francisella tularensis osteomyelitis of the
biol. 2000;74:179-193. hand following a cat bite: a case of clinical suspicion. Plast Recon-
6. Love DN, Jones RF, Bailey M, et al. Bacteria isolated from subcu- str Surg. 2011;128:37e-39e.
taneous abscesses in cats. Aust Vet Practit. 1978;8:87-90. 27. McCabe SJ, Murray JF, Ruhnke HL, et al. Mycoplasma infection of
7. Norris JM, Love DN. The isolation and enumeration of three feline the hand acquired from a cat. J Hand Surg Am. 1987;12:1085-1088.
oral Porphyromonas species from subcutaneous abscesses in cats. 28. Oehler RL, Velez AP, Mizrachi M, et al. Bite-related and septic syn-
Vet Microbiol. 1999;65:115-122. dromes caused by cats and dogs. Lancet Infect Dis. 2009;9:439-447.
8. Malik R, Norris J, White J, et al. Wound cat. J Feline Med Surg. 29. Boinett C, Gonzalez A. Pasteurella multocida septicaemia in a
2006;8:135-140. patient on haemodialysis. BMJ Case Rep. 2009.
9. Costanzo C, Garosi LS, Glass EN, et al. Brain abscess in seven cats 30. Bryant BJ, Conry-Cantilena C, Ahlgren A, et al. Pasteurella multo-
due to a bite wound: MRI findings, surgical management and out- cida bacteremia in asymptomatic plateletpheresis donors: a tale of
come. J Feline Med Surg. 2011;13:672-680. two cats. Transfusion. 2007;47:1984-1989.
10. Fossum TW, Hedlund CS, Johnson AL, et al. Surgery of the integu- 31. Waghorn DJ, Robson M. Occupational risk of Pasteurella multo-
mentary system. In: Fossum TW, ed. Small Animal Surgery. 3rd ed. cida septicaemia and premature labour in a pregnant vet. BJOG.
St. Louis, MO: Mosby; 2007:159-259. 2003;110:780-781.
11. The Humane Society of the United States. 2012. http://www. 32. Wade T, Booy R, Teare EL, et  al. Pasteurella multocida men-
humanesociety.org/issues/pet_overpopulation/facts/pet_ownership_ ingitis in infancy-(a lick may be as bad as a bite). Eur J Pediatr.
statistics.html. Last accessed January 1, 2013. 1999;158:875-878.
12. Pet Population 2011. 2011. http://www.pfma.org.uk. Last accessed 33. Gaastra W, Lipman LJ. Capnocytophaga canimorsus. Vet Micro-
January 1, 2013. biol. 2010;140:339-346.
13. Bregman B, Slavinski S. Using emergency department data to con- 34. Pers C, Gahrn-Hansen B, Frederiksen W. Capnocytophaga cani-
duct dog and animal bite surveillance in New York City, 2003- morsus septicemia in Denmark, 1982-1995: review of 39 cases.
2006. Public Health Rep. 2012;127:195-201. Clin Infect Dis. 1996;23:71-75.
14. Talan DA, Citron DM, Abrahamian FM, et al. Bacteriologic analy- 35. de Smet MD, Chan CC, Nussenblatt RB, et  al. Capnocytophaga
sis of infected dog and cat bites. Emergency Medicine Animal Bite canimorsus as the cause of a chronic corneal infection. Am J
Infection Study Group. N Engl J Med. 1999;340:85-92. ­Ophthalmol. 1990;109:240-242.
15. Weiss HB, Friedman DI, Coben JH. Incidence of dog bite injuries 36. Chodosh J. Cat’s tooth keratitis: human corneal infection with
treated in emergency departments. JAMA. 1998;279:51-53. Capnocytophaga canimorsus. Cornea. 2001;20:661-663.
SECTION 3
Fungal and Algal Diseases

CHAPTER 58

Dermatophytosis
Jane E. Sykes and Catherine A. Outerbridge

other infected animals or through contact with contaminated


Overview of Dermatophytosis fomites (which includes the haircoats of animals and arthro-
First Described: Dermatophytosis was described indepen- pods such as fleas or houseflies). Dermatophyte spores survive
dently by several scientists in the 1830s and 1840s,1 but more than a year in the environment under optimal conditions
probable descriptions of ringworm date back to ancient of temperature and humidity, and they resist most routinely
Greek times. Dermatophytes were more fully character- used hospital disinfectants, which facilitates transmission. Der-
ized in the early 1900s by Sabouraud.2 matophytes are somewhat host species specific and are classified
Causes: Microsporum and Trichophyton species (especially as geophilic, zoophilic, or a­ nthropophilic (Table 58-1).3-8 Geo-
Microsporum canis, Microsporum gypseum, and Trichophy- philic dermatophytes are soil saprophytes. The most common
ton mentagrophytes) (teleomorph name, Arthroderma spp.) geophilic dermatophyte that infects dogs or cats is ­Microsporum
gypseum, which is most prevalent in warm, humid tropical and
Geographic Distribution: Worldwide, especially warm and
subtropical environments. Zoophilic dermatophytes are adapted
humid climates
to animal hosts and are rarely found in soil. The most common
Mode of Transmission: Direct contact with other infected zoophilic dermatophyte that infects dogs and especially cats is
hosts, fomite transmission Microsporum canis; this organism accounts for more than 90%
Major Clinical Signs: Cutaneous lesions include alopecia, ery- of dermatophyte isolates from cats worldwide and more than
thema, crusting and scaling, and sometimes nodular or 60% of isolates from dogs.3,9,10 Sylvatic dermatophytes are zoo-
draining lesions (kerions and dermatophytic mycetomas). philic dermatophytes that are adapted to rodents or hedgehogs.
The most common sylvatic dermatophyte that infects dogs and
Differential Diagnoses: Alopecic lesions must be differentiated
cats is Trichophyton ­mentagrophytes. Anthropophilic derma-
from self-traumatic alopecia secondary to allergic dermati-
tophytes are adapted to human hosts and do not survive in the
tis, sarcoptic mange, or demodicosis. If scale and crusting
soil; they include ­Microsporum ­audouinii, Trichophyton ton-
are also present, bacterial folliculitis, superficial pyoderma,
surans, ­Trichophyton rubrum, and Epidermophyton ­floccosum.
and pemphigus foliaceus are differential diagnoses. Nodu-
Rarely, these species infect or contaminate dogs or cats that
lar lesions must be differentiated from eosinophilic granu-
have a history of close contact with infected humans.4,11-16
loma, feline acne (cats), and mast cell tumors (dogs).
Mixed infections with multiple dermatophyte species occur
Human Health Significance: Dermatophytosis is a zoono- rarely in dogs.10
sis. Young children and the immunocompromised can Risk factors for dermatophytosis in dogs and cats include
develop extensive cutaneous lesions. young age and concurrent immunosuppressive disorders,
especially endogenous or iatrogenic hyperadrenocorticism.
Dermatophytosis is more common in cats than in dogs, but
Etiology and Epidemiology the prevalence varies with geographic location. Positive der-
matophyte cultures occurred in 6% of submissions from dogs
Dermatophytosis (ringworm or tinea) is a superficial cutaneous or cats in the southern United States (3.8% for dogs and
infection with one or more of the keratinophilic fungi that belong 14.9% for cats),17 and in the United Kingdom, 16% were pos-
to the genera Microsporum, Trichophyton, or E ­ pidermophyton. itive (10% for dogs and 16% for cats).9 Higher prevalences of
Transmission of dermatophytes occurs by close contact with detection (19% to 43%) have been reported in some studies

558
CHAPTER 58  Dermatophytosis 559

TABLE 58-1
Zoophilic and Geophilic Dermatophyte Species Reported to Infect Dogs and/or Cats3-8
Dermatophyte Species Classification Primary Reservoir Species Affected Geographic Distribution
Microsporum canis Zoophilic Cats, dogs, horses All mammals Temperate and tropical
locations worldwide
Microsporum gypseum Geophilic Soil All mammals Worldwide
Microsporum persicolor* Zoophilic Voles, field mice Dogs, cats Europe
Trichophyton mentagrophytes Zoophilic Rodents All mammals Worldwide
Trichophyton erinacei Zoophilic Hedgehogs Dogs Europe and New Zealand
Trichophyton simii Zoophilic Primates Dogs, cats India and the Far East
Trichophyton terrestre* Geophilic Soil All mammals Worldwide

*Do not invade hair; the role of T. terrestre in disease causation is controversial.

of cats and dogs, with and without skin lesions, from parts
of continental Europe.3,5,10 Dermatophytosis (especially geo-
philic dermatophytosis) is more prevalent in regions with high
warmth and humidity. Shelter cats, the vast majority of which
lack skin lesions, are more likely to carry dermatophytes on
their haircoats than healthy pet cats. Dermatophytes were
cultured from 5.5% and 19% of all shelter cats depend-
ing on geographical region,18,19 whereas the prevalence of
M. canis isolation from healthy pet cats is generally less than
2.5%.18 Animals admitted to breeding or boarding facili-
ties that have a history of dermatophytosis are also at risk
of infection, whereas dermatophytes are generally not detect-
able on the haircoats of animals in facilities without a his-
tory of infection.20 Retrovirus-infected cats are no more likely
to carry dermatophytes than retrovirus-negative cats,21 but
retrovirus-infected cats carry a greater range of species and
may be more likely to develop skin lesions (dermatophytosis).
Genetic factors may also be important. Persian and Himala-
yan cats and Yorkshire and Jack Russell terriers appear to
be predisposed to dermatophytosis.3,9,17 In particular, Persian
cats are thought to be at greater risk for development of der-
matophytic mycetomas (see Clinical Features). Dogs that hunt
A
and burrow in soil may be at increased risk for infection by
geophilic or sylvatic dermatophytes, which may explain the
common distribution of lesions on the face and distal thoracic
limbs for these dermatophyte species (Figure 58-1).

Clinical Features
Signs and Their Pathogenesis
Pathogenic dermatophytes invade keratinized tissue that has
been macerated or traumatized. Infections are localized to the
stratum corneum, hair shafts, and claws. The clinical expres-
sion of the disease is affected by the fungal species and strain
involved and the host response to infection.22 Infectious arthro-
spores, which are formed by the segmentation and fragmenta-
tion of fungal hyphae, adhere to the surfaces of skin and nails,
germinate, and secrete proteases. These proteases, such as sub-
tilisins, fungalysins, and a variety of acidic proteases,22,23 digest
keratin into short peptides or amino acids, which are then used B
by the fungus as a source of nutrition. The pattern of prote- FIGURE 58-1  Chronic Trichophyton mentagrophytes infections. There is alope-
ases secreted by different dermatophyte strains may influence cia, crusting, hyperpigmentation, and ulceration of the muzzle and periocular skin. 
the clinical expression of disease. On the host, dermatophytes A, Four-and-a-half-year-old male neutered fox terrier. B, Six-year-old female spayed Jack
reproduce asexually through production of conidia. Russell terrier. (Courtesy University of California, Davis, Veterinary Dermatology Service.)
560 SECTION 3  Fungal and Algal Diseases

A B
FIGURE 58-2  A, Jack Russell terrier puppy with multifocal classic well-circumscribed lesions caused by infection by Microsporum canis. B, On questioning, the owner of the affected
litter of puppies revealed that she also had a lesion on her arm. (Courtesy Dr. Terry Nagle.)

Longhaired cats may develop a poor haircoat, shed exces-


sively, and vomit hairballs more frequently, because infected
hair shafts are friable and prone to breakage. Persian and
Himalayan cats (and very rarely, domestic longhair cats and
dogs) with M. canis infections can develop dermatophytic
mycetomas, which are coalescing subcutaneous nodular
lesions that often drain purulent fluid (Figure 58-4).26-31 These
develop when dermatophytes invade the dermis and subcuta-
neous tissues, presumably after traumatic implantation of fun-
gal organisms from the haircoat of an already colonized or
infected animal. These lesions have also been referred to as
granulomatous dermatitis, Majocchi’s granuloma, or “pseu-
domycetomas,” although use of the last term was not recom-
mended by the International Society for Human and Animal
Mycology (ISHAM) committee for nomenclature of fungal
diseases.32 In contrast to true mycetomas (eumycetomas), der-
matophytic mycetomas are not caused by soil saprophytes, and
lesions are often multiple and coalescing, rather than single
and localized.10
FIGURE 58-3  Nine-year-old female spayed labrador retriever with multiple kerions Physical Examination Findings
on the muzzle due to Microsporum canis infection. Another dog in the household had
similar lesions that were less severe. (Courtesy University of California, Davis, Veterinary In dogs, dermatophytosis can occur anywhere on the body but
Dermatology Service.) most often occurs on the face, distal limbs, and tail. Dogs are
more likely than cats to present with classical, localized, well-
circumscribed lesions. Claw infection is manifested by erythema
Dermatophyte lesions form after an incubation period of 1 and thickening of the ungual fold, deformity and friability of the
to 3 weeks. Classically, lesions are single or multiple and consist claw, and sometimes footpad involvement (Figure 58-5). Kerions
of circular regions of alopecia with an erythematous margin, most commonly develop on the muzzle or distal limbs, and with
superficial scale and crust, and follicular papules and pustules digital pressure they typically have a boggy or exudative appear-
(Figure 58-2). Central healing may occur, sometimes with ance (see Figure 58-4).
hyperpigmentation or hair regrowth in the center of the lesion. Lesions in cats are extremely pleomorphic and can consist
Pruritis is variable. Infection by T. mentagrophytes in dogs of one or more areas of partial alopecia with scale and crusts,
can result in progressive alopecia, with dramatic scaling and/ especially on the pinnae and pinnal margins, nasal bridge, distal
or crusting and inflammation and in some cases scarring (see limbs, or tail (Figure 58-6). Some lesions resemble miliary der-
Figure 58-1). A kerion or kerion reaction (also known as nodu- matitis or are focal, pruritic lesions that resemble eosinophilic
lar dermatophytosis) is a localized, nodular inflammatory lesion plaques. Cats with dermatophytic mycetomas have focal (and
that results from dermatophyte infection and may be associated rarely disseminated) nodular, sometimes ulcerated cutaneous
with infection by M. gypseum, T. mentagrophytes, or M. canis lesions, usually on the dorsum or neck, which may drain serous
(Figure 58-3).10,24,25 or purulent fluid (see Figure 58-4).
CHAPTER 58  Dermatophytosis 561

Diagnosis abnormalities associated with predisposing disease condi-


tions (such as hyperadrenocorticism).
Diagnosis of dermatophytosis is based on clinical suspicion
together with the results of Wood’s lamp examination, skin and Microbiologic Tests
hair cytology, skin scrapings (to evaluate for demodicosis), and/or Wood’s Lamp Examination
fungal culture (Table 58-2). In both dogs and cats, lesions are rarely A positive Wood’s lamp examination is characterized by the pres-
symmetric. This asymmetry can help differentiate dermatophyto- ence of apple-green fluorescence along the length of a hair shaft
sis from other diagnoses that more commonly cause symmetrical (Figure 58-7). The fluorescence results from tryptophan metabo-
lesions. However, diagnosis that is solely based on the appearance lites produced by some M. canis strains when they grow within the
of lesions and/or Wood’s lamp examination is not recommended, hair shaft; the metabolite is not produced when the organism grows
because it can lead to misdiagnosis and unnecessary treatment that on scale or the claw. A significant proportion of M. canis infections
may be expensive and even harmful. In addition, mixed processes do not fluoresce,3,9 and other dermatophyte species of veterinary
can occur, such as co-infections with dermatophytes and staphy- significance do not produce fluorescent metabolites, so the sensi-
lococci or dermatophytes and Demodex mites. Because dermato- tivity of Wood’s lamp examination is low. The lamp should be
phytosis can take on a wide range of appearances in cats, it should allowed to warm up for several minutes before it is used, and the
be considered a possible differential diagnosis in cats with any skin lights should be turned off during the examination. False-positive
disease.33 results can occur if topical ointments, scale, lint, keratinized debris,
or sebum are present and incorrectly interpreted as dermatophyte
Laboratory Abnormalities fluorescence. Hairs that fluoresce can be plucked and submitted for
Complete Blood Count, Serum Biochemical Tests, and Urinalysis cytologic examination and fungal culture.
The results of routine laboratory testing in dogs and cats
with dermatophytosis are typically unremarkable or show Cytologic Examination
Cytologic examination of hairs plucked from lesions or the cir-
cumference of lesions may reveal fungal hyphae or chains of
arthroconidia (ectothrix spores) in some affected animals; mac-
roconidia, which form in culture, are not seen (Figure 58-8).
Addition of a clearing agent such as 10% or 20% potassium
hydroxide (KOH) or chlorphenolac is recommended, which
removes scale and debris that can interfere with organism iden-
tification. Potassium hydroxide solutions must be gently heated
with the hair for 10 minutes for clearing to occur, or incubated

B
FIGURE 58-5  Foot of a 9-year-old intact male terrier mix with dermatophyto-
FIGURE 58-4  A and B, Twelve-year-old female spayed Himalayan with a derma- sis caused by Microsporum gypseum. The dog competed in rodent hunting events that
tophytic mycetoma. Ulceration and fistulation are present. Ultimately, surgical excision occurred in underground tunnels. Severe, chronic dermatitis with ulceration and alopecia
of the mycetoma was required. (Courtesy University of California, Davis, Veterinary that involved multiple distal limbs and claws was present. The claws were irregular and
Dermatology Service.) discolored. (Courtesy University of California, Davis, Veterinary Dermatology Service.)
562 SECTION 3  Fungal and Algal Diseases

overnight at room temperature. Chlorphenolac, which is com-


posed of chloral hydrate, phenol, and lactic acid, is not readily
available and is toxic, but it does not require heating or pro-
longed incubation. Examination of hairs mounted in mineral oil
can still yield positive results in the absence of a clearing agent.25
Cytologic examination has low sensitivity when compared
with culture, and experience is required for proper organism
identification.

Fungal Culture
Culture is the gold standard for diagnosis of dermatophytosis.
Ten to 12 hairs (ideally those that appear damaged or broken)
should be plucked from the edge of lesions with sterile forceps
and placed in a clean, dry paper envelope (preferably) or a ster-
ile tube or container at room temperature. Scrapings of crusts
from the edge of lesions or nail clippings (after cleaning the nail
A
with 70% alcohol) can also be submitted (see also Chapter 4 for
specimen collection methods for dermatophytes). Alternatively,
a brand-new toothbrush (out of the package) can be used to
collect specimens from lesions or the entire coat of a suspect ani-
mal by vigorous brushing for several minutes. The toothbrush
is then submitted to the laboratory. Macerated tissue cultures
can be performed on biopsy specimens from animals that have
nodular dermatophytosis or dermatophytic mycetomas.
The most commonly used media for culture are Dermato-
phyte Test Medium (DTM) or Sabouraud’s dextrose agar. In-
house DTM cultures, if performed, should be interpreted with
caution. Flat Petri plates (as opposed to vials) should be used in
order to facilitate inoculation and species identification. Inocu-
lated media should be incubated at 24°C to 30°C; incubation at
room temperature is more likely to result in false negatives.34
Growth occurs within 14 days, and often within 5 to 7 days,
but may take up to 21 days (particularly for treated animals).
Dermatophyte colonies, which are white or cream and powdery
(Figure 58-9), should not be confused with those of saprophytic
molds, which may be brown, green, black, gray, or mixed in
B color. The DTM turns red when the first dermatophyte colony
is visible. Color changes that occur after the first colony appears
FIGURE 58-6  Eighteen-year-old male neutered domestic shorthair cat with dermato-
phytosis caused by Trichophyton mentagrophytes. The paws (A), pinnae (B), and nasal bridge may result from growth of saprophytic fungi. All suspicious col-
were involved. (Courtesy University of California, Davis, Veterinary Dermatology Service.) onies must be confirmed as dermatophytes and speciated based

TABLE 58-2
Diagnostic Assays Available for Dermatophytosis
Assay Specimen Type Target Performance
Wood’s lamp Haircoat Fluorescent Low sensitivity (around 50%) for detection of M. canis when
examination Microsporum performed properly. Other dermatophyte species do not fluo-
canis strains in resce. The presence of scale, sebum, debris, and topical oint-
hair ments may lead to false positive results. When positive, allows
identification of suitable specimens for cytology and/or culture.
Cytology Hair pluck specimens Dermatophyte When positive, allows early institution of treatment. Low
hyphae and sensitivity. Incubation with clearing agents may increase
arthrospores sensitivity, but these may not be readily available. Other
fungal conidia that contaminate the haircoat or debris may
be mistaken for dermatophyte elements.
Culture Plucked hair specimens, All dermatophyte Gold standard for diagnosis of dermatophytosis. Prolonged
scale and nail clippings species incubation times may be required.
Histopathology Skin biopsies Dermatophytes Organisms may be visualized with silver stains, periodic
acid–Schiff stain, or immunohistochemistry.
CHAPTER 58  Dermatophytosis 563

on spore morphology through the use of microscopy (see Figure


58-9). Spores form after 7 to 10 days of growth. The individual
performing the identification should wear gloves. Tape is placed
over the colony and transferred to a slide, on which a drop of
stain is applied. A coverslip is then applied. Lactophenol cotton
blue or new methylene blue are suitable stains. Dysgonic strains
of M. canis have been described that have atypical morphology
in culture, but these are rare.27,35 Whenever possible, submis-
sion to a veterinary diagnostic laboratory that follows Clinical
and Laboratory Standards Institute (CLSI) guidelines is rec-
ommended to ensure proper quality assurance for culture and
identification of dermatophytes. This also minimizes the risk of
laboratory-acquired infections and contamination of the prac-
tice environment. Matrix-assisted laser desorption/ionization–
time of flight mass spectrometry (MALDI-TOF) offers promise
for rapid identification of dermatophytes in veterinary diagnos-
tic microbiology laboratories in the future.36

FIGURE 58-7  Positive Wood’s lamp examination in a 2-year-old male neutered Molecular Diagnosis Using the Polymerase Chain Reaction
Persian that was seen for pruritis and hair loss. (Courtesy University of California, Davis, PCR assays have been developed that detect and differentiate
Veterinary Dermatology Service.)
between dermatophytes that infect dogs and cats.37,38 Sensitive
real-time PCR assays have been described for use in humans,39,40
but their use has not been reported for the diagnosis of derma-
tophytosis in dogs or cats. PCR assays have the potential to be

A B

C
FIGURE 58-8  Cytology images of hairs infected with dermatophytes. A, Unstained preparation. Large numbers of Microsporum canis spores are seen that surround the hair shaft.
B, Parker ink-KOH stain. Arthrospores and hyphae can be seen. C, Unstained preparation of a hair infected by Microsporum gypseum.
564 SECTION 3  Fungal and Algal Diseases

more sensitive than culture, with a more rapid turnaround time, and PAS-positive fungal elements that are embedded in an
and have the potential to facilitate assessment of treatment effi- eosinophilic amorphous material within the dermis.
cacy and control of disease in group-housed animals.
Treatment and Prognosis
Pathologic Findings
Histopathology of biopsy specimens from dogs or cats with Most otherwise healthy animals with dermatophytosis clear the
dermatophytosis may reveal spores and hyphae within intrafol- infection without specific antifungal treatment within 3 months of
licular hair shafts, in association with a variable pyogranuloma- diagnosis. Nevertheless, treatment is strongly recommended for
tous inflammatory response (Figure 58-10). Special stains such animals that reside in multianimal households and those that live
as periodic acid–Schiff (PAS) or Gomori’s methenamine silver with immunocompromised humans (which includes children and
can increase the sensitivity for detection of fungal elements.41 older people, e.g., 60+ years of age) because of the highly conta-
Immunohistochemical stains have also been used to detect gious nature of the infection. The general approach to treatment
M. canis within lesions.26 Occasionally, dermatophytes produce includes the use of systemic antifungal drugs, with or without
a superficial pustular histologic lesion. Acantholytic keratino- topical treatment, together with environmental decontamination.
cytes can be seen in biopsies from some dogs, which may lead Lesions should be mapped and photographed before treatment is
to misdiagnosis of pemphigus foliaceus.41 Histopathology of initiated so that progress can be monitored over time.
representative skin biopsies is especially useful for diagnosis Management of dermatophytosis in multicat households
of dermatophytic mycetomas, kerions, and fungal paronychia, can be frustrating, because (1) all in-contact cats must be
because organisms are typically present in low numbers in these treated; (2) some cats are colonized and have no visible lesions;
lesions and culture may be negative. Histopathology of derma- and (3) testing and treatment require considerable energy,
tophytic mycetomas reveals pyogranulomatous inflammation time, monitoring, and expense. For group-housed animals, all
in-contact animals should be examined and cultured using the
toothbrush technique. Ideally all animals with positive cultures
(and definitely those with clinical signs) should be treated with
both systemic and topical antifungal therapy, whereas all unaf-
fected, culture-negative animals should be monitored for devel-
opment of lesions and treated with topical antifungal drugs
(ideally lime sulfur).

Topical Antifungal Drug Treatment


Topical antifungal drug treatment is not as effective as systemic
treatment, but inactivates fungal spores on the haircoat and
reduces environmental contamination. Spot treatments are not
recommended, because spores are distributed all over the hair-
coat even when skin lesions are localized. The two most effective
topical treatments are lime sulfur (1:16 dilution, or 237 mL in
3.6 L of water) or 0.2% enilconazole, both of which should be
applied twice weekly.42-44 Miconazole-chlorhexidine shampoos
are also an option but may not be as effective as lime sulfur.45,46
Lime sulfur shampoos, sprays, or dips have a foul odor and can

B
FIGURE 58-9  Selected cultural characteristics of dermatophytes. A, Microsporum FIGURE 58-10  Histopathology that shows a cross-sectional view of a hair shaft
canis from a cat with a dermatophytic mycetoma growing on inhibitory mold agar. from a 3-year-old female spayed Boston terrier. Fungal hyphae are visible within the hair
B, Macroconidia of M. gypseum after growth in culture. shaft (arrows).
CHAPTER 58  Dermatophytosis 565

TABLE 58-3
Antimicrobial Drugs That Can Be Used for Systemic Treatment of Dermatophytosis*
Drug Dose (mg/kg) Route Interval (hours)
Itraconazole† 5 PO 12-24
Fluconazole 5-10 PO 12-24
Ketoconazole 10 PO 12
Terbinafine† 30-40 PO 24
Griseofulvin† (microsized) 25 PO 12
Griseofulvin (ultramicrosized) 15 PO 12

*See also Chapter 9 for more information on adverse reactions and drug administration before use.
†Administer itraconazole capsules, terbinafine, and griseofulvin with food. Reduce dose of itraconazole to 3 mg/kg q24h if using the suspension in cats.

stain light-colored haircoats but are safe for all age groups. A Environmental Decontamination
reformulated lime sulfur product with an odor-masking agent In some situations, environmental decontamination is extremely
was not as effective in one clinical trial as a traditional lime difficult or impossible because of the hardiness of dermatophyte
sulfur preparation.46 Lime sulfur products are most commonly spores and their widespread dispersal on surfaces that cannot
used in the United States but may not be readily available in be adequately disinfected. Even concentrated (1:10) bleach solu-
other parts of the world. Anecdotally, lime sulfur solutions have tions are not 100% active against dermatophyte spores; com-
been reported to cause oral ulcerations in cats, but these were plete disinfection requires undiluted household bleach or 1%
not detected in studies that evaluated cats that were allowed formalin. Both are too toxic and caustic for routine use.
to lick their coats after treatment.46,47 Ingestion of enilconazole All potentially contaminated beds and blankets should be
can cause hypersalivation, anorexia, and vomiting, and there either discarded or washed with bleach through the use of a hot
are rare anecdotal reports of severe adverse effects that include cycle and a hot dryer. Leashes, collars, and grooming equipment
death.11 Use of an Elizabethan collar is recommended to prevent should be discarded, and carpets and furniture should be vacu-
cats from licking topical solutions from the haircoat until the umed to remove all visible hair and then steam cleaned. Because
treatment has dried. Chlorhexidine alone or povidone-iodine are vacuum cleaners are a source of recontamination, they may need
not sufficiently active against spores and are not recommended. to be replaced after initial cleaning efforts; at the very least the
Whether or not the haircoat should be clipped before applica- vacuum bag should be discarded. Disinfection of hard surfaces
tion of these treatments is controversial. Clipping removes con- should be performed with 1:10 to 1:100 bleach solution, 0.2%
taminated hair and facilitates penetration of topical treatments, enilconazole solution, or (where available) an enilconazole fog-
but may also serve to contaminate the environment and may ger. Vehicles and carriers used to transport animals must also be
create or exacerbate skin lesions if clipping disrupts the skin. It appropriately cleaned. According to label information, concen-
is recommended that clipping be used for longhaired cats and all trated (1:16) accelerated hydrogen peroxide solutions may also
cats in an affected Persian or Himalayan cattery. Clippers must inactivate dermatophytes and reduce environmental contamina-
be thoroughly disinfected with 10% bleach or concentrated tion, but this requires further evaluation (see Chapter 11).
accelerated hydrogen peroxide solutions afterward.
Duration of Treatment
Systemic Antifungal Drug Treatment The duration of treatment of dogs and cats with dermatophy-
Systemic antifungal drug treatment decreases the duration tosis is based on serial physical examinations and importantly,
and severity of feline dermatophytosis (Table 58-3).42 How- follow-up culture results. Usually, several months of treatment
ever, it should be used in conjunction with topical treatment are required. In general, animals should be examined and a cul-
and environmental decontamination when groups of animals ture performed at least monthly (and no more frequently than
are affected. Antifungal drugs with activity against dermato- weekly) during treatment. Treatment should be continued until
phytes are griseofulvin, ketoconazole, fluconazole, itraconazole, two successive cultures are negative and active lesions have
and terbinafine (see Chapter 9 for additional information on resolved. For group-housed animals, at least three successive
these drugs). Itraconazole is the drug of choice for treatment negative cultures are suggested. Some authors advocate once-
of dermatophytosis, because adverse reactions are uncommon weekly cultures because this may permit earlier discontinua-
and it has equal or greater efficacy when compared with gris- tion of treatment.11 If serial cultures are not possible because
eofulvin.48 Fluconazole is less active than other azole antifungal of financial limitations, treatment should be continued for 2 to
drugs against dermatophytes in  vitro (as is the case for other 4 weeks after complete resolution of clinical signs. Typically,
fungi),49,50 but controlled clinical trials that compare the efficacy treatment is continued for at least 10 to 12 weeks.
of fluconazole to that of itraconazole for treatment of dermato-
phytosis are needed. The chitin inhibitor lufenuron (traditionally Prognosis
administered orally to prevent flea infestations) has been evalu- The prognosis is good for resolution of dermatophytosis in
ated for treatment of dermatophytosis,51,52 but its efficacy has young animals, especially in households with only a single
been questioned and its use is no longer recommended.10,11,24 animal or very low numbers of animals. Complicated disease
566 SECTION 3  Fungal and Algal Diseases

such as dermatophytic mycetomas are difficult to resolve and


require very long treatment durations; in some cats with myce-
tomas, surgery may ultimately be required for complete lesion
resolution. The prognosis for rapid resolution of dermatophy-
tosis may also be worse for animals with irreversible underlying
immunocompromise. Hair regrowth may not occur in regions
of scarring.

Immunity and Vaccination


Resolution of dermatophytosis depends on a delayed type-
hypersensitivity immune response, which involves the activity
of macrophages and IFN-γ.22 Less invasive dermatophytes may
evade the host immune response by limiting their presence to
the superficial corneal epithelial cells and hair shafts. A vaccine
for prevention of feline M. canis infections was licensed in the
United States in the mid-1990s but was not sufficiently effica-
cious and is no longer available.

Prevention
Prevention of dermatophytosis relies on avoidance of contact
with infected or carrier animals or fomites, but this may be
impossible in some situations. For scenarios that involve group-
housed animals, clean (culture-negative) animals, incoming ani-
mals, and animals undergoing treatment (culture-positive) must
be kept in separate rooms, and ideally in individual cages. Once
animals in the contaminated room are culture negative, they
are moved to the clean room, and the contaminated room is
thoroughly disinfected. Culture and topical treatment should be
considered for any incoming animals to a clean facility. FIGURE 58-11  Boy with multiple lesions of dermatophytosis on the face and body.
In veterinary hospitals, animals with known dermatophy- The boy had juvenile rheumatoid arthritis. The boy’s cat had a lesion on the right side of its
tosis should be scheduled for examination late in the day and face. (Courtesy Dr. Richard Malik.)
should be carried directly from the parking lot to an appropri-
ate examination room that can subsequently be easily cleaned
(the animal should not wait in the reception area). Strict atten- tinea capitis in the United States, with most cases caused by
tion to barrier precautions is required (see Chapter 11). If hos- anthropophilic dermatophytes such as T. tonsurans.56
pitalization is necessary, animals with suspected or documented Young children and the immunocompromised (e.g., trans-
M. canis infections should be housed in isolation. plant recipients) are most susceptible to dermatophytosis and
may develop deep, recalcitrant, and/or disseminated lesions
Public Health Aspects as a result of contact with infected pets.57,58 Disease has been
described in veterinarians and veterinary students.11,35 In general,
Forms of human dermatophytosis include tinea capitis (scalp), zoophilic and geophilic species evoke a more profound inflamma-
tinea pedis (“athlete’s foot”), tinea barbae (bearded area), tory response than anthropophilic dermatophytes; the latter tend
tinea cruris (“jock itch”), tinea corporis (body), tinea imbri- to cause more chronic infections in people.1 Typical “ringworm”
cata (a variant of tinea corporis), tinea manuum (hand), tinea lesions in humans are erythematous, circular lesions with a raised
faciei (face), and tinea unguium (nails).1 Dermatophyte spe- margin (Figure 58-11), but atypical forms that mimic other skin
cies that cause disease in dogs and cats have the potential to diseases such as atopic dermatitis and lupus have been described.59
cause human disease. Microsporum canis is the most common Humans who handle infected animals should wear protective
zoophilic dermatophyte implicated. Zoophilic dermatophytes clothing and gloves. Young children or the immunocompromised
are most often associated with tinea capitis or tinea corporis. should avoid any contact with these animals. Toothbrush cul-
A high prevalence of infection with M. canis has been reported tures should be considered for cats that are adopted from shelters
in children that have tinea capitis (scalp infection) from con- or multicat households before they are introduced to households
tinental Europe and Asia.53-55 M. canis is now a rare cause of where immunocompromised individuals reside.
CHAPTER 58  Dermatophytosis 567

CASE EXAMPLE Eyes, Ears, and Throat: No clinically significant abnormalities


of the eyes or ears were noted. On oral examination, the
dentition was severely worn, and there was mild to moderate
Signalment: “Jack”, a 4.5-year-old male neutered smooth- periodontal disease.
coated fox terrier from northern California Musculoskeletal: Normal ambulation. Body condition score
History: Jack was evaluated for chronic diarrhea, a distended was 3/9.
abdomen, and loss of hair on the face. Jack’s owners adopted Cardiovascular and Respiratory Systems: No clinically
him when he was 14 months old, at which time he had no significant abnormalities were noted.
clinical signs of illness. At 2 years of age, the owners noted that Gastrointestinal and Genitourinary: The abdomen was
Jack began to rub his muzzle on objects. The intensity of this moderately distended with a palpable fluid wave. Rectal
behavior at times resulted in excoriations that would bleed. examination revealed only a moderate amount of formed
Jack had been seen at another veterinary clinic where he was but soft feces.
treated with a diet that was “hypoallergenic.” The owners then Lymph Nodes: No peripheral lymphadenopathy was detected.
continued to try to feed a variety of different diets for the skin Laboratory Findings:
problem, with a change to a new diet approximately every CBC:
3 months. The most recently fed diets were a prescription HCT 39.8% (40%-55%)
intestinal diet and a prescription venison and potato diet. Diet MCV 69.7 fL (65-75 fL)
trials were never strict elimination trials, because the dog was MCHC 34.2 g/dL (33-36 g/dL)
fed the same treats throughout the diet trial process. There WBC 18,810 cells/µL (6000-13,000 cells/µL)
was no improvement in the skin lesions, which progressed to Neutrophils 16,177 cells/µL (3000-10,500 cells/µL)
cause alopecia that involved the entire aspect of his face (see Band neutrophils 188 cells/µL
Figure 58-1, A). Approximately 18 months before Jack was Lymphocytes 376 cells/µL (1000-4000 cells/µL)
evaluated, intermittent diarrhea had developed. The diarrhea Monocytes 1693 cells/µL (150-1200 cells/µL)
was brown, occurred once or twice a day, and was sometimes Eosinophils 376 cells/µL (0-1500 cells/µL)
accompanied by tenesmus. It occasionally contained Platelets clumped, rare nucleated RBC/100 WBC.
mucus but never blood, and varied from soft to watery in Serum Chemistry Profile:
consistency. In addition to this history, over the preceding Sodium 144 mmol/L (145-154 mmol/L)
2 years, Jack had been treated intermittently with prednisone Potassium 4.1 mmol/L (3.6-5.3 mmol/L)
(0.5 to 0.75 mg/kg PO q12h) for the pruritus. The prednisone Chloride 115 mmol/L (108-118 mmol/L)
never improved the skin condition but was occasionally Bicarbonate 24 mmol/L (16-26 mmol/L)
associated with resolution of the diarrhea. Other medications Phosphorus 1.9 mg/dL (3.0-6.2 mg/dL)
used at various time points over the previous 2 years included Calcium 6.6 mg/dL (9.7-11.5 mg/dL)
hydroxyzine, metronidazole, tylosin, cephalexin, and a topical BUN 10 mg/dL (5-21 mg/dL)
otic preparation that contained thiabendazole, neomycin, Creatinine 0.5 mg/dL (0.3-1.2 mg/dL)
and dexamethasone. Although Jack’s appetite was generally Glucose 108 mg/dL (64-123 mg/dL)
good, he was described as a picky eater and had intermittent Total protein 2.9 g/dL (5.4-7.6 g/dL)
lethargy when his diarrhea was severe. The owners reported Albumin 1.5 g/dL (3.0-4.4 g/dL)
that over the most recent few weeks he had lost weight and Globulin 1.4 g/dL (1.8-3.9 g/dL)
muscle mass and had developed a distended abdomen. Jack ALT 85 U/L (19-67 U/L)
lived on a ranch with multiple other dogs (the owner bred AST 109 U/L (19-42 U/L)
poodles and shelties), cats, and horses, none of which were ALP 23 U/L (21-170 U/L)
unwell or had skin lesions. He spent most of his time outdoors Creatine kinase 309 U/L (51-399 U/L)
on the farm and loved to dig in rodent holes. GGT < 3 U/L (0-6 U/L)
Current medications: Prednisone, 1.4 mg/kg PO q12h Cholesterol 88 mg/dL (135-361 mg/dL)
Physical Examination: Total bilirubin < 0.1 mg/dL (0-0.2 mg/dL)
Body Weight: 7.1 kg. Magnesium 1.8 mg/dL (1.5-2.6 mg/dL).
General: Bright, alert, responsive, hydrated. T = 100.3°F Serum Vitamin B12 and Folate: Vitamin B12 314 ng/L (272-875
(37.9°C), HR = 120 beats/min, RR = 40 breaths/min, mucous ng/L), folate 9.1 ng/mL (6.5-18.6 ng/mL).
membranes pink/pigmented, CRT <2 s. Serum Ionized Magnesium Concentration: 0.44 mmol/L
Integument: A dull, unkempt haircoat was noted. An area of (0.40-0.52 mmol/L).
partial alopecia with crusts was present over the right caudal Urinalysis (Cystocentesis): SGr 1.018; pH 6.5, no protein,
tarsus. Diffuse, complete alopecia with serocellular and bilirubin, hemoprotein, or glucose, 0-2 WBC/HPF, no RBC/
hemorrhagic crusting and multifocal, discrete ulcerations HPF, no crystals or bacteria, rare granular casts, few lipid
was present over the entire dorsal muzzle (see Figure 58-1, A). droplets.
The skin within the alopecic area was hyperpigmented and Imaging Findings:
had a shiny appearance compatible with scarring. Periocular Thoracic Radiographs: No abnormalities were detected in
alopecia with crusts was also present bilaterally and was the thorax. In the portion of visible abdomen, there was
confluent with the erythematous, advancing edge of the decreased serosal detail.
facial lesion. The skin on the ventral abdomen was thin with Abdominal Ultrasound: There was marked thickening of
prominent subcutaneous vessels. the mucosal layer of the small intestine diffusely, with
568 SECTION 3  Fungal and Algal Diseases

hyperechoic striations. A moderate volume of anechoic Physical examination revealed resolution of crusts and
peritoneal effusion was present. evidence of new hair growth. Body weight had reduced to
Cytologic Findings: Abdominal effusion: the fluid was 6.2 kg and no evidence of abdominal distention was present.
colorless with a total protein of 0.3 g/dL, low numbers of The only abnormalities on the chemistry panel were mild
erythrocytes, and 380 nucleated cells/µL (38% neutrophils, hypoproteinemia (5.1 g/dL) and hypocholesterolemia (110
2% small mononuclear cells, and 60% large mononuclear mg/dL). Albumin was 3.1 g/dL and globulin was 2.2 g/dL.
cells). Nucleated cells were a mixture of nondegenerate Abdominal ultrasound examination showed resolution of
neutrophils and mononuclear cells of normal morphology. ascites. The prednisone dose was decreased to 0.4 mg/kg
One large cluster of reactive mesothelial cells was observed. PO q24h, but diarrhea returned, so the dose was increased
The fluid was interpreted as a transudate. again to 0.4 mg/kg PO q12h. A repeat fungal culture (hair
Microbiologic Testing: Skin scrapings and hair pluck pluck specimen) 3 months after starting treatment was
cytology (trichogram) (from face and tarsus): Large numbers negative; culture was not performed earlier because lesions
of dermatophyte hyphae and spores were present. No mites still appeared active, even though they had improved.
were seen. Additional follow-up showed complete resolution of
Fungal culture (hair specimens from periphery of lesions on abnormalities on the serum chemistry panel and serum B12
face and tarsus): Small numbers of Trichophyton mentagro- and folate concentrations in the middle of the reference
phytes grew after 4 days. range. Ultimately management of the diarrhea required a
Diagnosis: Protein-losing enteropathy (suspect lymphangiec- cottage cheese and rice–based diet (balanced by a veterinary
tasia), likely iatrogenic hyperadrenocorticism, and dermato- nutritionist) and a low dose of prednisone (0.4 mg/kg PO
phytosis due to T. mentagrophytes infection. every 4 days). However, despite resolution of crusts and
Treatment: Before the results of the fungal culture became pruritis, hair cultures continued to periodically grow small
available, fluconazole (7 mg/kg PO q24h) was prescribed numbers of T. mentagrophytes 1 year later, and fluconazole
based on the high index of suspicion from cytologic was never discontinued.
identification of fungal elements, together with an oatmeal Comments: Persistent dermatophytosis in this dog was
shampoo to be used twice weekly. The dose of prednisone likely due to underlying immunosuppressive disease/drug
was reduced to 0.5 mg/kg PO q12h and strict instructions treatment that could not be completely reversed, and
were given to feed a prescription low-fat diet and no other possibly reinfection. The dog was likely exposed as a result
foods (including treats). At a recheck 3 weeks later, the of digging in rodent holes, based on the history and the
owners reported that Jack was doing very well. His diarrhea, dermatophyte species isolated. Poor client compliance and
abdominal distention, and facial pruritus had resolved; client financial limitations were factors that may also have
he was more energetic; and his appetite had improved. contributed to an inability to control the infection.

SUGGESTED READINGS 9. Sparkes AH, Gruffydd-Jones TJ, Shaw SE, et  al. Epidemiological
and diagnostic features of canine and feline dermatophytosis in the
Chermette R, Ferreiro L, Guillot J. Dermatophytoses in animals. Myco- United Kingdom from 1956 to 1991. Vet Rec. 1993;133:57-61.
pathologia. 2008;166:385-405. 10. Chermette R, Ferreiro L, Guillot J. Dermatophytoses in animals.
Moriello KA. Treatment of dermatophytosis in dogs and cats: review of Mycopathologia. 2008;166:385-405.
published studies. Vet Dermatol. 2004;15:99-107. 11. Moriello K, DeBoer DJ. Cutaneous fungal infections. In: Greene
Vermout S, Tabart J, Baldo A, et al. Pathogenesis of dermatophytosis. CE, ed. Infectious Diseases of the Dog and Cat. 4th ed. St Louis,
Mycopathologia. 2008;166:267-275. MO: Elsevier; 2012:588-606.
12. Kushida T, Watanabe S. Canine ringworm caused by Trichophyton
REFERENCES rubrum; probable transmission from man to animal. Sabouraudia.
1. Weitzman I, Summerbell RC. The dermatophytes. Clin Microbiol 1975;13(Pt 1):30-32.
Rev. 1995;8:240-259. 13. Kano R, Hirai A, Yoshiike M, et  al. Molecular identification of
2. Sabouraud R. Les teignes. Paris: Masson et Cie; 1910. Trichophyton rubrum isolate from a dog by chitin synthase 1 (CHS1)
3. Cafarchia C, Romito D, Sasanelli M, et  al. The epidemiology of gene analysis. Med Mycol. 2002;40:439-442.
canine and feline dermatophytoses in southern Italy. Mycoses. 14. Brilhante RS, Cordeiro RA, Gomes JM, et al. Canine dermatophy-
2004;47:508-513. tosis caused by an anthropophilic species: molecular and phenotyp-
4. Ranganathan S, Arun Mozhi Balajee S, Mahendra Raja S. A survey ical characterization of Trichophyton tonsurans. J Med Microbiol.
of dermatophytosis in animals in Madras, India. Mycopathologia. 2006;55:1583-1586.
1997;140:137-140. 15. Terreni AA, Gregg Jr WB, Morris PR, et al. Epidermophyton floc-
5. Seker E, Dogan N. Isolation of dermatophytes from dogs and cats cosum infection in a dog from the United States. Sabouraudia.
with suspected dermatophytosis in Western Turkey. Prev Vet Med. 1985;23:141-142.
2011;98:46-51. 16. Stenwig H, Taksdal T. Isolation of Epidermophyton floccosum
6. Muller A, Guaguere E, Degorce-Rubiales F, et al. Dermatophytosis from a dog in Norway. Sabouraudia. 1984;22:171-172.
due to Microsporum persicolor: a retrospective study of 16 cases. 17. Lewis DT, Foil CS, Hosgood G. Epidemiology and clinical features
Can Vet J. 2011;52:385-388. of dermatophytosis in dogs and cats at Louisiana State University:
7. Fairley RA. The histological lesions of Trichophyton mentagrophytes 1981-1990. Vet Dermatol. 1991;2:53-58.
var erinacei infection in dogs. Vet Dermatol. 2001;12:119-122. 18. Mignon BR, Losson BJ. Prevalence and characterization of
8. Pierard-Franchimont C, Hermanns JF, Collette C, et al. Hedgehog Microsporum canis carriage in cats. J Med Vet Mycol. 1997;35:
ringworm in humans and a dog. Acta Clin Belg. 2008;63:322-324. 249-256.
CHAPTER 58  Dermatophytosis 569

19. Moriello KA, Kunkle G, DeBoer DJ. Isolation of dermatophytes 41. Peters J, Scott DW, Erb HN, et al. Comparative analysis of canine
from the hair coats of stray cats from selected animal shelters in dermatophytosis and superficial pemphigus for the prevalence of
two different geographic regions of the United States. Vet Derma- dermatophytes and acantholytic keratinocytes: a histopathological
tol. 1994;5:57-62. and clinical retrospective study. Vet Dermatol. 2007;18:234-240.
20. Moriello KA, Deboer DJ. Fungal flora of the haircoat of cats with 42. Moriello KA. Treatment of dermatophytosis in dogs and cats:
and without dermatophytosis. J Med Vet Mycol. 1991;29:285-292. review of published studies. Vet Dermatol. 2004;15:99-107.
21. Reche Jr A, Daniel AG, Lazaro Strauss TC, et al. Cutaneous myco- 43. Diesel A, Verbrugge M, Moriello KA. Efficacy of eight commer-
flora and CD4:CD8 ratio of cats infected with feline immunodefi- cial formulations of lime sulphur on in vitro growth inhibition of
ciency virus. J Feline Med Surg. 2010;12:355-358. Microsporum canis. Vet Dermatol. 2011;22:197-201.
22. Vermout S, Tabart J, Baldo A, et al. Pathogenesis of dermatophyto- 44. Hnilica KA, Medleau L. Evaluation of topically applied enilcon-
sis. Mycopathologia. 2008;166:267-275. azole for the treatment of dermatophytosis in a Persian cattery. Vet
23. Sriranganadane D, Waridel P, Salamin K, et  al. Identification of Dermatol. 2002;13:23-28.
novel secreted proteases during extracellular proteolysis by derma- 45. Moriello KA, Verbrugge M. Use of isolated infected spores to
tophytes at acidic pH. Proteomics. 2011;11:4422-4433. determine the sporocidal efficacy of two commercial antifungal
24. Outerbridge CA. Mycologic disorders of the skin. Clin Tech Small rinses against Microsporum canis. Vet Dermatol. 2007;18:55-58.
Anim Pract. 2006;21:128-134. 46. Newbury S, Moriello KA, Kwochka KW, et al. Use of itraconazole
25. Cornegliani L, Persico P, Colombo S. Canine nodular dermatophy- and either lime sulphur or Malaseb Concentrate Rinse (R) to treat
tosis (kerion): 23 cases. Vet Dermatol. 2009;20:185-190. shelter cats naturally infected with Microsporum canis: an open
26. Abramo F, Vercelli A, Mancianti F. Two cases of dermatophytic field trial. Vet Dermatol. 2011;22:75-79.
pseudomycetoma in the dog: an immunohistochemical study. Vet 47. Newbury S, Moriello K, Verbrugge M, et al. Use of lime sulphur
Dermatol. 2001;12:203-207. and itraconazole to treat shelter cats naturally infected with Micros-
27. Thian A, Woodgyer AJ, Holloway SA. Dysgonic strain of Micros- porum canis in an annex facility: an open field trial. Vet Dermatol.
porum canis pseudomycetoma in a domestic long-hair cat. Aust Vet 2007;18:324-331.
J. 2008;86:324-328. 48. Moriello KA, DeBoer DJ. Efficacy of griseofulvin and itraconazole
28. Nobre Mde O, Negri Mueller E, Teixeira Tillmann M, et al. Dis- in the treatment of experimentally induced dermatophytosis in cats.
ease progression of dermatophytic pseudomycetoma in a Persian J Am Vet Med Assoc. 1995;207:439-444.
cat. Rev Iberoam Micol. 2010;27:98-100. 49. Perea S, Fothergill AW, Sutton DA, et al. Comparison of in vitro
29. Bergman RL, Medleau L, Hnilica K, et al. Dermatophyte granulo- activities of voriconazole and five established antifungal agents
mas caused by Trichophyton mentagrophytes in a dog. Vet Derma- against different species of dermatophytes using a broth macrodilu-
tol. 2002;13:49-52. tion method. J Clin Microbiol. 2001;39:385-388.
30. Chang SC, Liao JW, Shyu CL, et al. Dermatophytic pseudomyceto- 50. Mota CR, Miranda KC, Lemos Jde A, et al. Comparison of in vitro
mas in four cats. Vet Dermatol. 2011;22:181-187. activity of five antifungal agents against dermatophytes, using the
31. Bond R, Pocknell AM, Tozet CE. Pseudomycetoma caused by agar dilution and broth microdilution methods. Rev Soc Bras Med
Microsporum canis in a Persian cat: lack of response to oral terbin- Trop. 2009;42:250-254.
afine. J Small Anim Pract. 2001;42:557-560. 51. Guillot J, Malandain E, Jankowski F, et al. Evaluation of the efficacy
32. Odds FC, Arai T, Disalvo AF, et al. Nomenclature of fungal dis- of oral lufenuron combined with topical enilconazole for the manage-
eases: a report and recommendations from a Sub-Committee of the ment of dermatophytosis in catteries. Vet Rec. 2002;150:714-718.
International Society for Human and Animal Mycology (ISHAM). 52. Ben-Ziony Y, Arzi B. Use of lufenuron for treating fungal infections
J Med Vet Mycol. 1992;30:1-10. of dogs and cats: 297 cases (1997-1999). J Am Vet Med Assoc.
33. Chermette R, Bussieras S, Jeanmonod P, et  al. Dermatophytie à 2000;217:1510-1513.
Trichophyton rubrum chez un chien et son propriétaire. Première 53. Tsoumani M, Jelastopulu E, Bartzavali C, et al. Changes of derma-
description en France. Bull Soc Fr Mycol Med. 1990;19:219-223. tophytoses in southwestern Greece: an 18-year survey. Mycopatho-
34. Guillot J, Latie L, Deville M, et al. Evaluation of the dermatophyte logia. 2011;172:63-67.
test medium RapidVet-D. Vet Dermatol. 2001;12:123-127. 54. del Boz J, Crespo V, Rivas-Ruiz F, et al. A 30-year survey of paedi-
35. Hermoso de Mendoza M, Hermoso de Mendoza J, Alonso JM, et al. atric tinea capitis in southern Spain. J Eur Acad Dermatol Venereol.
A zoonotic ringworm outbreak caused by a dysgonic strain of Micros- 2011;25:170-174.
porum canis from stray cats. Rev Iberoam Micol. 2010;27:62-65. 55. Zhu M, Li L, Wang J, et al. Tinea capitis in Southeastern China: a
36. Nenoff P, Erhard M, Simon JC, et al. MALDI-TOF mass spectrom- 16-year survey. Mycopathologia. 2010;169:235-239.
etry—a rapid method for the identification of dermatophyte species. 56. Coloe JR, Diab M, Moennich J, et al. Tinea capitis among children
Med Mycol. 2013;51:17-24. in the Columbus area, Ohio, USA. Mycoses. 2010;53:158-162.
37. Cafarchia C, Gasser RB, Figueredo LA, et al. An improved molecu- 57. Trabelsi S, Aouinet A, Abderrahim E, et al. First case of subcutane-
lar diagnostic assay for canine and feline dermatophytosis. Med ous dermatomycoses in a Tunisian renal transplant patient. Tunis Med.
Mycol. 2013;51:136-143. 2012;90:196-199.
38. Nardoni S, Franceschi A, Mancianti F. Identification of Micros- 58. Chiapello LS, Dib MD, Nuncira CT, et al. Mycetoma of the scalp
porum canis from dermatophytic pseudomycetoma in paraffin- due to Microsporum canis: hystopathologic, mycologic, and immu-
embedded veterinary specimens using a common PCR protocol. nogenetic features in a 6-year-old girl. Diagn Microbiol Infect Dis.
Mycoses. 2007;50:215-217. 2011;70:145-149.
39. Yuksel T, Ilkit M. Identification of rare macroconidia-producing der- 59. Atzori L, Aste N, Pau M. Tinea faciei due to Microsporum canis
matophytic fungi by real-time PCR. Med Mycol. 2012;50:346-352. in children: a survey of 46 cases in the district of Cagliari (Italy).
40. Jensen RH, Arendrup MC. Molecular diagnosis of dermatophyte Pediatr Dermatol. 2012;29:409-413.
infections. Curr Opin Infect Dis. 2012;25:126-134.
CHAPTER 59

Malassezia Infections
Jane E. Sykes, Terry M. Nagle, and Stephen D. White

ears of dogs and cats with otitis externa than from the ears of
Overview of Malassezia Dermatitis healthy dogs and cats.6,7 Dogs with skin disease can have 100-
First Described: 1874, in France, by Louis-Charles Malassez to 10,000-fold increases in skin population densities of Malas-
(the fungus was initially named Pityrosporum ovale)1 sezia spp. compared with healthy dogs.8 Species and strains of
Cause: Malassezia pachydermatis; less commonly, lipid- Malassezia vary in virulence and tropism for different anatomic
dependent species such as Malassezia globosa, Malasse- sites and host species.9,10 A variety of lipid-dependent Malasse-
zia sympodialis, Malassezia nana, Malassezia slooffiae, and zia spp. occasionally have been isolated from dogs and cats with
Malassezia furfur are involved. otitis externa.6,7,11-14
Malassezia spp. infection can occur in dogs and cats of any
Affected Hosts: Dogs and to a lesser extent cats
age, breed, and sex, but some dog breeds are thought to be pre-
Geographic Distribution: Worldwide disposed, including American cocker spaniels, West Highland
Mode of Transmission: Direct contact and opportunistic pro- white terriers, basset hounds, poodles, and Australian silky terri-
liferation of commensal organisms ers.15 Sphynx and Devon Rex cats have high rates of Malassezia
spp. colonization when compared with domestic shorthair cats.16
Major Clinical Signs: Cutaneous erythema, pruritus, alopecia,
scaling, greasy exudation, hyperpigmentation, lichenifi- Clinical Features
cation, malodor
Differential Diagnoses: Dermatophytosis, demodicosis, aller- Signs and Their Pathogenesis
gic dermatitis, drug eruptions, pyoderma Malassezia spp. proliferate opportunistically, becoming patho-
Human Health Significance: M. pachydermatis does not nor- genic with alterations in host defenses or the skin surface
mally colonize human skin but can rarely infect immuno- microclimate. Examples of underlying diseases include allergic
suppressed adults and cause catheter-related infections dermatitis, endocrinopathies, intertrigo, primary keratinization/
in neonatal intensive care units. cornification disorders, and, in cats, underlying neoplasia or ret-
rovirus infections.17,18 A history of treatment with antibiotics
was a predisposing factor in one study.19 Prolonged glucocor-
ticoid treatment can also lead to yeast proliferation. The yeasts
Etiology and Epidemiology adhere to the cells of the stratum corneum and secrete lipases,
proteinases, phospholipases, and acid sphingomyelinases.20
Malassezia spp. (formerly Pityrosporum spp.) are lipophilic The resulting skin lesions may be localized or generalized, are
yeasts that normally colonize animal and human skin in low thought to result from inflammation, and, in some animals,
numbers. At least 13 species of Malassezia exist. The vast major- hypersensitivity reactions to yeast antigens.21,22 The more ery-
ity of species are classified as lipid dependent: when grown in thematous the interdigital region is in an atopic dog, the more
the laboratory, these species have an absolute requirement for likely a Malassezia spp. infection is present.23
long-chain fatty acids, which are used as a source of carbon. The most commonly affected anatomic sites are the ear
In contrast, Malassezia pachydermatis, the most commonly canals, skin folds (including the periocular and perioral skin,
isolated yeast species from the skin and ears of healthy dogs ventral neck, axillae, inguinal regions, and perineum), and
and cats, is non–lipid dependent when grown in the laboratory. interdigital skin (Figure 59-1). Clinical signs include ery-
Lipid-dependent species, including Malassezia globosa, Malas- thema, pruritus, alopecia, scaling, and a greasy exudate. The
sezia sympodialis, Malassezia nana, Malassezia slooffiae, and claw beds may develop a reddish-brown stain. Chronicity
Malassezia furfur, can colonize the skin of healthy cats.2-5 results in hyperpigmentation, lichenification, and stenosis of
In healthy dogs, Malassezia spp. are most commonly found the ear canal, and a pungent, offensive odor develops. Pruritus
in the ear canals, on the lips, axillae, interdigital spaces, anal may result in excoriations. Devon Rex and Sphynx cats can
sacs, and occasionally the nose and vagina. It has been suggested develop a greasy dermatitis associated with increased numbers
that a symbiotic relationship exists between Malassezia spp. and of Malassezia spp., which can respond dramatically to anti-
commensal staphylococci; each maintains a local microenviron- fungal drug treatment.8
ment that benefits the other.
Malassezia spp. are common opportunistic contributors to Physical Examination
chronic dermatitis and otitis externa in dogs, and to a lesser A thorough physical and dermatologic examination aids diag-
extent in cats. They are isolated more frequently from the nosis of Malassezia spp. infection and provides important clues

570
CHAPTER 59  Malassezia Infections 571

A B
FIGURE 59-1  Five-year-old male neutered pug with a 1 year history of otitis externa and generalized pruritus. Malassezia dermatitis and bacterial pyoderma secondary to underlying
atopic dermatitis was diagnosed. Note the distribution of lesions, which affect the facial folds and ventral neck (A) and axillae (B).

as to the underlying disorder leading to opportunistic prolifera-


tion of Malassezia spp. It also allows identification of lesions
consistent with concurrent pyoderma (see Chapter 84).

Diagnosis
Diagnosis of Malassezia spp. dermatitis is based on clinical signs,
findings on cytologic examination, and a positive response to
antifungal drug treatment.

Microbiologic Testing
Skin Scrapings
Skin scrapings are important in animals suspected to have Mal-
assezia spp. dermatitis in order to rule out demodicosis, which
can mimic Malassezia spp. dermatitis.

Cytologic Examination
Cytology is an important tool for diagnosis and is generally
performed using clear (not frosted) acetate (Scotch) tape prepa- FIGURE 59-2  Tape preparation from the skin of a dog with Malassezia dermatitis.
Note broad-based budding yeasts, which have the appearance of a footprint. Modified
rations of the skin. The tape is pressed to the skin, dipped in
Wright’s stain, 1000× oil magnification.
the final step basophilic (blue) Diff-Quik solution, rinsed, and
applied to a slide on which a drop of immersion oil has been
placed. Alternatively, the tape is pressed against the affected into account other factors, such as strain variation in virulence
skin several times and then placed on a dry glass slide, and a or host hypersensitivity.10,24 For Malassezia spp. dermatitis, mean
small amount of the basophilic Diff-Quik solution is injected counts of 1 or more yeast per oil immersion field (1000× magnifi-
under the tape. The tape is then examined using a light micro- cation) are considered abnormal in dogs.25 For ears, a mean yeast
scope, with an additional drop of immersion oil to allow count of approximately 5 or more organisms per oil immersion
identification of bacteria and Malassezia spp. using the 100˜ field has been suggested as abnormal.25 Cats may be colonized
objective. The yeasts stain deeply basophilic and exhibit wide- with higher numbers of yeasts, and 12 or more organisms per
based budding, resembling “footprints,” “peanuts,” or “snow- high dry field has been suggested as abnormal in feline ear canals
men” (Figure 59-2). An inflammatory cellular response is often in another study.26 The ear canals of healthy dogs and cats have
not present, even when Malassezia spp. contribute to disease mean counts of 2 or fewer yeasts per high dry field (400× magnifi-
progression. cation), and intermediate counts represent a gray zone.26
Estimation of yeast counts, although only semiquantitative,
may help to determine the role of Malassezia in otitis and der- Fungal Culture
matitis, but there are no clear guidelines as to what constitutes a Fungal culture and susceptibility testing for Malassezia spp.
normal yeast population size. In addition, yeast counts do not take is not routinely performed for clinical diagnostic purposes,
572 SECTION 3  Fungal and Algal Diseases

TABLE 59-1
Systemic Antifungal Drugs Used for Treatment of Malassezia Dermatitis and Otitis Externa in Dogs
Drug Dose (mg/kg) Route Interval (hours) Duration (days)
Ketoconazole 5 to 10 PO 12 3 to 4 weeks or until
Itraconazole 5* PO 24 cytologic resolution
Fluconazole 5 PO 24
Terbinafine 30 PO 24

*Dose applies to the capsules. Reduce dose to 3 mg/kg if using the suspension.

although methods that use broth dilution or E-testing for sus- Administration of lipid-containing total parenteral nutrition
ceptibility testing have been described. The organism can be solutions is a risk factor for disseminated infections in humans,
cultured from a tape preparation, which in the laboratory is which often occur in neonates.33,34
mounted on a drop of olive oil that has been placed on fungal M. pachydermatis is not part of the normal human cutane-
isolation media.25 Growth occurs within 7 days of incubation. ous microflora. Human disease associated with M. pachyder-
Currently, most Malassezia spp. infections are broadly suscep- matis infection occurs rarely. Nosocomial infections with M.
tible and respond well to treatment of the yeast infection and pachydermatis have been reported in neonates in intensive care
any underlying disorders present. One azole-resistant isolate of units.35 One epidemic was associated with colonization of health
M. pachydermatis was identified in a dog with dermatitis, but care workers’ pet dogs with a strain identical to that causing
this was not associated with resistance in vivo.27 disease, as determined by molecular typing with pulsed-field gel
electrophoresis.36 Cutaneous infections have also been reported
Treatment and Prognosis in adults with underlying immunosuppressive disease.30 Owners
of dogs with atopic skin disease were 11 times more likely to
Treatment of Malassezia spp. dermatitis and otitis requires iden- carry M. pachydermatis on their hands than owners of healthy
tification and, if possible, treatment of the underlying cause, in dogs, as determined using culture (39% vs. 6%).25 The owners
addition to antifungal drug treatment. Because Malassezia spp. of both groups of dogs were equally likely to carry the yeast on
infection is superficial, topical treatments may be adequate to their hands as determined by PCR testing (93% and 94%). It
resolve infection. A variety of topical otic preparations are avail- was concluded that mechanical carriage of the organism by dog
able for treatment of Malassezia otitis. These contain antifun- owners appears to be of low risk to human health, given how
gals such as clotrimazole, miconazole, or posaconazole, which rarely disease due to M. pachydermatis occurs. Hand washing
are generally combined with a glucocorticoid and an antibacte- is likely to reduce the rate of carriage of M. pachydermatis, but
rial (see Chapter 84). There is good evidence that Malassezia this has not been well studied.
dermatitis can be successfully treated with twice-weekly sham-
pooing with a 2% miconazole/2% chlorhexidine shampoo (e.g.,
Malaseb, DVM Pharmaceuticals) for 3 weeks,24 and this can
also prevent recurrence. A 3% chlorhexidine shampoo may be
equally efficacious.28 Systemic antifungals that have been shown CASE EXAMPLE
to have efficacy for treatment of Malassezia infections include
ketoconazole, itraconazole, fluconazole, and terbinafine, which See case example in Chapter 84.
are generally administered for 3 weeks (Table 59-1).29,30 Longer
periods of treatment may be required for severe infections or
infections of the claw bed.8 Administration of terbinafine on
two consecutive days each week (i.e., in a pulsatile fashion)
SUGGESTED READING
may be as effective as daily administration, but more studies Tragiannidis A, Bisping G, Koehler G, et al. Minireview: Malassezia infec-
are required.31 Treatment should be monitored through serial tions in immunocompromised patients. Mycoses. 2010;53(3):187-195.
dermatologic examinations and cytologic evaluation of tape
REFERENCES
preparations. The prognosis for resolution depends on the abil-
ity to identify and address the underlying cause for proliferation 1. Malassez L. Note sur la champignon de la pelade. Arch Physiol
of Malassezia spp. Norm et Pathol. 1874;1:203-212.
2. Bond R, Anthony RM, Dodd M, et al. Isolation of Malassezia sym-
podialis from feline skin. J Med Vet Mycol. 1996;34(2):145-147.
Public Health Aspects 3. Bond R, Howell SA, Haywood PJ, et  al. Isolation of Malassezia
sympodialis and Malassezia globosa from healthy pet cats. Vet Rec.
In human beings, Malassezia infections most commonly result 1997;141(8):200-201.
from infection by M. furfur. This organism belongs to the 4. Crespo MJ, Abarca ML, Cabañes FJ. Isolation of Malassezia furfur
normal human cutaneous flora and proliferates opportunisti- from a cat. J Clin Microbiol. 1999;37(5):1573-1574.
cally following immunocompromise. Yeast proliferation can 5. Volk AV, Belyavin CE, Varjonen K, et al. Malassezia pachydermatis
lead to Malassezia folliculitis, seborrheic dermatitis, catheter- and M. nana predominate amongst the cutaneous mycobiotia of
related fungemia, and a variety of other invasive infections.32 Sphynx cats. J Fel Med Surg. 2010;12(12):917-922.
CHAPTER 59  Malassezia Infections 573

6. Cafarchia C, Gallo S, Capelli G, et al. Occurrence and population 23. White SD, Bourdeau P, Blumstein P, et al. Comparison via cytology
size of Malassezia spp. in the external ear canal of dogs and cats both and culture of carriage of Malassezia pachydermatis in atopic and
healthy and with otitis. Mycopathologica. 2005;160(2):143-149. healthy dogs. In: Kwochka KW, Willemse T, von Tscharner C, eds.
7. Dizotti CE, Coutinho SD. Isolation of Malassezia pachydermatis Advances in Veterinary Dermatology. vol. 3. Oxford, UK: Butter-
and M. sympodialis from the external ear canal of cats with and worth Heinemann; 1998:292-298.
without otitis externa. Acta Vet Hung. 2007;55(4):471-477. 24. Negre A, Bensignor E, Guillot J. Evidence-based veterinary derma-
8. Bond R. Superficial veterinary mycoses. Clin Dermatol. 2010;28(2): tology: a systematic review of interventions for Malassezia derma-
226-236. titis in dogs. Vet Dermatol. 2009;20(1):1-12.
9. Duarte ER, Hamdan JS. RAPD differentiation of Malassezia spp. 25. Morris DO. Malassezia pachydermatis carriage in dog owners.
from cattle, dogs and humans. Mycoses. 2010;53(1):48-56. Emerg Infect Dis. 2005;11(1):83-88.
10. Machado ML, Cafarchia C, Otranto D, et  al. Genetic variability 26. Ginel PJ, Lucena R, Rodriguez JC, et  al. A semiquantitative
and phospholipase production of Malassezia pachydermatis iso- cytological evaluation of normal and pathological samples
lated from dogs with diverse grades of skin lesions. Med Mycol. from the external ear canal of dogs and cats. Vet Dermatol.
2010;48(6):889-892. 2002;13(3):151-156.
11. Cafarchia C, Latrofa MS, Figueredo LA, et  al. Physiological and 27. Nijima M, Kano R, Nagata M, et al. An azole-resistant isolate of
molecular characterization of atypical lipid-dependent Malassezia Malassezia pachydermatis. Vet Microbiol. 2011;149:288-290.
yeasts from a dog with skin lesions: adaptation to a new host? Med 28. Maynard L, Rème CA, Viaud S. Comparison of two shampoos for
Mycol. 2011;49:365-374. the treatment of Malassezia dermatitis: a randomised controlled
12. Crespo MJ, Abarca ML, Cabaûes FJ. Otitis externa associ- trial. J Small Anim Pract. 2011;52:566-572.
ated with Malassezia sympodialis in two cats. J Clin Microbiol. 29. Rosales MS, Marsella R, Kunkle G, et al. Comparison of the clini-
2000;38(3):1263-1266. cal efficacy of oral terbinafine and ketoconazole combined with
13. Crespo MJ, Abarca ML, Cabaûes FJ. Atypical lipid-dependent cephalexin in the treatment of Malassezia dermatitis in dogs—a
Malassezia species isolated from dogs with otitis externa. J Clin pilot study. Vet Dermatol. 2005;16(3):171-176.
Microbiol. 2000;38(6):2383-2385. 30. Sickafoose L, Hosgood G, Snook T, et al. A noninferiority clinical
14. Shokri H, Khosravi A, Rad M, et al. Occurrence of Malassezia spe- trial comparing fluconazole and ketoconazole in combination with
cies in Persian and domestic short hair cats with and without otitis cephalexin for the treatment of dogs with Malassezia dermatitis.
externa. J Vet Med Sci. 2010;72(3):293-296. Vet Ther. 2010;11(2):E1-E13.
15. Scott DW, Miller WH, Griffin CE. Bacterial skin diseases. In: Muller 31. Berger DJ, Lewis TP, Schick AE, et al. Comparison of once-daily
and Kirk’s Small Animal Dermatology. 6th ed. Philadelphia, PA: versus twice-weekly terbinafine administration for the treatment
WB Saunders; 2001:274-335. of canine Malassezia dermatitis – a pilot study. Vet Dermatol.
16. Volk AV, Belyavin CE, Varjonen K, et al. Malassezia pachyderma- 2012;23:418–e79.
tis and M. nana predominate amongst the cutaneous mycobiota of 32. Tragiannidis A, Bisping G, Koehler G, et  al. Minireview: Mal-
Sphynx cats. J Fel Med Surg. 2010;2(12):917-922. assezia infections in immunocompromised patients. Mycoses.
17. Sierra P, Guillot J, Jacob H, et al. Fungal flora on cutaneous and 2010;53(3):187-195.
mucosal surfaces of cats infected with feline immunodeficiency 33. Dankner WM, Spector SA, Fierer J, et al. Malassezia fungemia in
virus or feline leukemia virus. Am J Vet Res. 2000;61(2):158-161. neonates and adults: complication of hyperalimentation. Rev Infect
18. Perrins N, Gaudiano F, Bond R. Carriage of Malassezia spp. yeasts Dis. 1987;9(4):743-753.
in cats with diabetes mellitus, hyperthyroidism and neoplasia. Med 34. Chryssanthou E, Broberger U, Petrini B. Malassezia pachyder-
Mycol. 2007;45(6):541-546. matis fungaemia in a neonatal intensive care unit. Acta Paediatr.
19. Plant JD, Rosenkrantz WS, Griffin CE. Factors associated with and 2001;90(3):323-327.
prevalence of high Malassezia pachydermatis numbers on dog skin. 35. Larocco M, Dorenbaum A, Robinson A, et  al. Recovery of Mal-
J Am Vet Med Assoc. 1992;201(6):879-882. assezia pachydermatis from eight infants in a neonatal intensive
20. Coutinho SD, Paula CR. Proteinase, phospholipase, hyaluronidase care nursery: clinical and laboratory features. Pediatr Infect Dis J.
and chondroitin-sulphatase production by Malassezia pachyderma- 1988;7(6):398-401.
tis. Med Mycol. 2000;38(1):73-76. 36. Chang HJ, Miller HL, Watkins N, et  al. An epidemic of Malas-
21. Bond R, Curtis CF, Hendricks A, et  al. Intradermal test reac- sezia pachydermatis in an intensive care nursery associated with
tivity to Malassezia pachydermatis in atopic dogs. Vet Rec. colonization of health care workers’ pet dogs. N Engl J Med.
2002;150(14):448-449. 1998;338(11):706-711.
22. Kim HJ, Kim ET, Lim CY, et al. The immunoglobulin G response
to Malassezia pachydermatis extracts in atopic and non-atopic
dogs. Can Vet J. 2010;51(8):869-872.
CHAPTER 60

Blastomycosis
Jane E. Sykes and Lindsay K. Merkel

the Ohio and Mississippi river valleys, and in the southeastern


Overview of Blastomycosis states (Figure 60-2). A smaller focus of endemicity exists in the
First Described: 1894, in the United States by Thomas Casper St. Lawrence river region in the northeast, which extends up
Gilchrist.1 The organism was initially mistaken for a proto- into Ontario, Canada. Disease may also be seen in parts of Can-
zoan parasite. ada around the Great Lakes and has also been reported from
Saskatchewan.2,3 Occasionally disease appears in nontraveled
Cause: Blastomyces dermatitidis (an ascomycete, teleomorph
dogs that reside in non-endemic parts of the United States, such
Ajellomyces dermatitidis)
as in Wyoming and South Dakota. Canine blastomycosis has
Affected Hosts: Primarily humans and dogs, but also other also been reported from India.4 Human blastomycosis occurs in
mammalian species that include cats, horses, ferrets, and Africa, but a different serotype is involved. Interestingly, reports
sea lions of canine disease from Africa are absent from the literature.
Geographic Distribution: Primarily North America, especially Blastomycosis was described in Europe in a dog with a travel
the south-central and upper midwestern states of the history to the United States.5
United States; Canadian provinces that border the Great The ecologic niche of B. dermatitidis appears to be warm, moist,
Lakes; and a small area of the northeastern United States sandy soil that is rich in organic debris such as decaying vegeta-
and southeastern Canada near the St. Lawrence river tion. However, this has not been thoroughly defined because the
fungus is difficult to isolate from the environment. No consistent
Mode of Transmission: Inhalation of conidia from the envi-
seasonality has been linked to disease. Genetic differences have
ronment; rarely cutaneous inoculation
been reported among isolates of B. dermatitidis within the United
Major Clinical Signs: Fever, inappetence, weight loss, cough, States. Using PCR-based typing methods, several different geno-
tachypnea, respiratory difficulty, nodular or ulcerative types have been described (A, B, C, D, and E).6 Analysis by mic-
cutaneous lesions, ocular lesions (uveitis, chorioretinitis, rosatellite typing suggests the existence of at least two genetically
panophthalmitis), lameness, neurologic signs distinct groups, group 1 and group 2, the geographic distributions
Differential Diagnoses: Neoplasia, other deep mycoses, pro- of which overlap.7 In human patients, group 2 isolates may be
tothecosis, mycobacterial infections, systemic protozoal more likely to be associated with disease in older patients with
infections (leishmaniasis, toxoplasmosis, neosporosis), comorbid conditions, and group 2 isolates are also more likely to
aspiration or foreign body pneumonia be associated with dissemination than group 1 isolates.8
Young adult, large-breed (>15 kg) dogs are predisposed
Human Health Significance: B. dermatitidis causes disease in to blastomycosis, and a slight male predisposition has been
humans, but direct transmission from animals to humans
does not occur (with the exception of rare bite wound
transmission from infected dogs). Dogs are a sentinel for
human exposure.

Etiology and Epidemiology


Blastomyces dermatitidis is a dimorphic fungus that grows as a
mycelial form in the environment and as a thick-walled budding
yeast in tissues (at 37°C). Hyphae within soil produce conidia
that are 2 to 10 µm in diameter. The conidia are thought to
be aerosolized and inhaled by the host, where they transform Lung
into yeasts and produce a localized pulmonary or disseminated
disease known as blastomycosis (Figure 60-1). The organism’s
Blood
name (dermatitidis) stems from the fact that the skin is a com-
mon site to which the organism disseminates. FIGURE 60-1  Life cycle of Blastomyces dermatitidis. From the lung, the organism
B. dermatitidis is primarily found in the eastern parts of can disseminate to a variety of tissues, but especially the eye, lymph nodes, skin, and
the United States, especially around the Great Lakes, along bones.

574
CHAPTER 60  Blastomycosis 575

TABLE 60-1
Physical Examination Findings in 115 Dogs with Blastomycosis9
Clinical Sign Percent of Dogs
Fever 62
Lymphadenopathy 56
Harsh lung sounds 50
Skin lesions 49
Chorioretinitis 43
Anterior uveitis 42
Cough 32
FIGURE 60-2  Approximate geographic distribution of Blastomyces dermatitidis in the
United States. Emaciation 25
Retinal detachment 23
Cutaneous mass 16
Glaucoma 16
identified in some studies.9 Overrepresented breeds include
coonhounds, pointers, Weimaraners, Labrador retrievers, Tachypnea 16
golden retrievers, and Doberman pinschers.9,10 This may reflect Dehydration 15
the increased likelihood of exposure of these dog types to organ- Bony mass/swelling 14
isms in the environment, because they are more likely to be used
Nasal discharge  8
in outdoor activities such as hunting. Dogs younger than 6
months of age and as old as 17 years may be affected, but dogs in Neurologic signs  6
the 2- to 4-year age group have the highest risk of infection with Prostatomegaly  5
the 4- to 6-year age group a close second.9,10 Residence less than Mammary mass  3
400 m from a body of water increased the risk of blastomycosis
Orchitis  2
by a factor of 10 in one study from Louisiana,10 and exposure to
sites of soil disturbance or excavation may also increase the risk Synovial effusion  2
of infection. In one report, B. dermatitidis was isolated from a
woodpile near the Wisconsin River; over 14 years, 4 of 9 dogs
housed in a kennel close to the woodpile developed blastomyco-
sis.11 Most affected dogs are immunocompetent. Blastomycosis virtually any tissue in the body may be affected, and infection
is rarely reported in cats from endemic regions of the United has been reported in the mammary glands, nasal or oral cavities,
States and, as with other soil-borne mycoses such as histoplas- cardiac tissues (myocardium, endocardium, and pericardium),
mosis and cryptococcosis, blastomycosis can occur in cats that and very rarely in the kidneys, liver, spleen, peritoneum, gastro-
have been housed entirely indoors.12,13 In one study, 10% of 41 intestinal tract, and urinary bladder.20
affected cats tested positive for FeLV antigen.14 Clinical signs are variable. Dogs may develop subclini-
cal infections, acute or chronic disease that is localized to the
Clinical Features lungs and associated lymph nodes, or severe and progressive
signs that result from dissemination of the organism to multiple
Signs and Their Pathogenesis extrapulmonary sites. The incubation period is variable and not
Infection most often results from inhalation of conidia in the precisely known, but is estimated to range from 5 to 12 weeks.
environment. Direct inoculation of organisms followed by Nonspecific signs of illness such as lethargy, weakness, fever,
localized cutaneous disease and/or osteomyelitis (inoculation inappetence, and weight loss are common (Table 60-1). Other
blastomycosis) occurs rarely in dogs and human patients.15,16 common clinical manifestations (between 20% and 50% of
Once within the host, the conidia transform into yeasts that can affected dogs) include signs of respiratory involvement (such as
resist destruction by neutrophils. The yeasts trigger a pyogranu- cough and increased respiratory rate), lymphadenopathy, ocular
lomatous inflammatory response. Important virulence factors manifestations, cutaneous lesions, and lameness.10,21 Lameness
include a cell surface glycoprotein known as BAD-1 (previously may result from fungal osteomyelitis, arthritis, or rarely, hyper-
known as WI-1), and other cell wall components such as trophic osteopathy.22 Gastrointestinal signs (vomiting, diarrhea,
α-1,3-glucan and possibly melanin.17,18 BAD-1 is an adhe- hematemesis, and melena); polyuria or polydipsia; mammary
sin that binds to host cell receptors on macrophages. It may gland masses; laryngeal masses; nasal discharge, sneezing and/
also contribute to the yeast’s ability to evade the host immune or epistaxis; testicular masses; dysuria or hematuria secondary
response by influencing cytokine secretion and impairing com- to prostatic involvement; cardiac arrhythmias; and neurologic
plement activation.17,19 In some animals, the organism spreads signs due to meningoencephalitis or ependymitis occur less fre-
hematogenously from the lungs to associated lymph nodes and quently.10,23-29 Neurologic involvement secondary to extension
other extrapulmonary sites. Extrapulmonary sites of predilec- of intranasal or retrobulbar Blastomyces granulomas through the
tion include the skin, eye, bone, reproductive tissues (prostate calvarium can also occur.25,29 Rarely, thromboembolic disease
and testes), and the central nervous system (CNS). However, has been reported.30
576 SECTION 3  Fungal and Algal Diseases

B C
FIGURE 60-3  Disseminated blastomycosis in a labrador retriever. Nodular cutaneous lesions are present on the muzzle (A). There are also multiple, ulcerated, and draining skin lesions
on the distal limbs (B) and digit (C). (Courtesy Dr. Sheila Torres, University of Minnesota.)

In cats, clinical signs of blastomycosis are similar to those


in dogs, although neurologic and gastrointestinal involvement
may be more prevalent.31

Physical Examination Findings


Physical examination abnormalities in dogs with blastomy-
cosis commonly include fever, thin body condition, dehydra-
tion, and signs of respiratory involvement. Fever is present in
approximately 50% of affected dogs and is usually low grade
(103°F to 104°F or 39.4°C to 40.0°C) but occasionally exceeds
106°F (41.1°C).20,27 Respiratory signs include cough, tachy-
pnea, increased respiratory effort, and increased lung sounds
on thoracic auscultation. Other common findings on physical
examination are firm cutaneous or subcutaneous masses, drain-
ing or ulcerated skin lesions, and peripheral lymphadenopathy.
Careful palpation of the entire skin surface can reveal small
skin lesions. Cutaneous and subcutaneous lesions are frequently
found on the trunk, limbs, and digits and can also involve the
muzzle (Figure 60-3). Lesions of the tongue, gingiva, or muco-
cutaneous junctions may also be present (Figure 60-4). Facial
swelling, exophthalmos, or facial deformity has been described
in some affected dogs.25,32 Dogs with nasal cavity involve-
ment can have stertorous respiration, decreased nasal airflow,
or nasal discharge. Ocular signs may be unilateral or bilateral
and include chorioretinitis (sometimes with retinal detach-
ment); lesions consistent with optic neuritis; endophthalmitis
or panophthalmitis; uveitis with aqueous flare, iris bombé, syn- FIGURE 60-4  Nodular and ulcerated lesions of the tongue in a dog with dissemi-
echia, and miosis; cataract formation; conjunctivitis; keratitis; nated blastomycosis. (Courtesy Dr. Sheila Torres, University of Minnesota.)
CHAPTER 60  Blastomycosis 577

A B

C
FIGURE 60-5  Cutaneous lesions in a cat with disseminated blastomycosis that involved the footpads (A), interdigital folds (B), and lips (C). (Courtesy Dr. Sheila Torres, University
of Minnesota.)

and photophobia.33,34 Severe ocular involvement may result in ataxia, pelvic limb paresis, circling, hyperesthesia, decreased
increased intraocular pressures and/or loss of vision. Dogs with placing reactions, and blindness.13,31,35-37
musculoskeletal involvement may be lame, have firm swellings
associated with long bones, or exhibit joint swelling, warmth, Diagnosis
and/or pain. Dogs with testicular blastomycosis may have scro-
tal swelling or palpable testicular masses. Neurologic signs are A diagnosis of blastomycosis is usually suspected based on
present in fewer than 5% of affected dogs and can include sei- the presence of suspicious clinicopathologic abnormalities in a
zures, hypermetria, decreased placing reactions, tetraparesis, dog or cat from an endemic area. The diagnosis is usually con-
circling, ataxia, blindness, decreased or absent menace and/ firmed through cytologic examination of fine needle aspirates
or pupillary light reflexes, nystagmus, and decreased facial of affected tissues (especially skin lesions and lymph nodes),
sensation. impression smears of draining skin lesions, respiratory lavage
Physical examination findings in cats with blastomycosis are specimens (transtracheal washes or bronchoalveolar lavage), or
similar to those described in dogs. These include fever, thin body body fluids (especially CSF or ocular fluid, but also synovial
condition, peripheral lymphadenopathy, tachypnea, increased fluid or urine sediment). Other methods of diagnosis include
or decreased lung sounds, ocular abnormalities (chorioretinitis histopathology (e.g., of bone or tissue biopsies), fungal culture,
with retinal detachment, uveitis, panophthalmitis, and second- and PCR-based assays (Table 60-2). Serologic assays that detect
ary glaucoma), nodular or draining skin lesions (Figure 60-5), antigen and antibody are also available, but when used alone,
and a variety of neurologic signs that include obtundation, these do not confirm a diagnosis of blastomycosis.
578 SECTION 3  Fungal and Algal Diseases

TABLE 60-2
Diagnostic Assays Currently Available for Blastomycosis in Dogs and Cats
Assay Specimen Type Target Performance
Cytologic Aspirates of affected tissues, Blastomyces dermatitidis Organisms are usually present in vari-
­examination body fluids, impression yeasts able numbers but occasionally may not
smears of skin lesions be visualized. Organisms may appear
disrupted after treatment with antifungal
drugs.
Fungal culture Aspirates or biopsies of B. dermatitidis Rarely indicated. Sensitive and specific
­affected tissues, body fluids and may be required for animals when
cytologic examination is negative. Risk
of laboratory-acquired infections. Slow
turnaround time (may require several
weeks of incubation).
Antibody serology Serum Antibodies to B. dermatitidis When present in conjunction with consis-
(gel ID) tent clinical signs, positive test results
usually indicate active infection, but
the potential for false positives exists.
Negative test results occur commonly in
dogs and cats with blastomycosis (low
sensitivity).
Antigen assay Urine, serum B. dermatitidis antigen Highly sensitive (>90%) when urine is used
(ELISA) as the test specimen. False positives are
extremely rare in dogs that lack fungal
disease, but cross-reactivity with other
fungal pathogens may occur (especially
Histoplasma), so positive results are not
diagnostic for blastomycosis.
Real-time PCR Whole blood B. dermatitidis DNA Not yet validated in adequate numbers of
assays dogs or cats with blastomycosis; useful-
ness requires further evaluation.
Histopathology Biopsy specimens from B. dermatitidis yeasts Sensitive, but yeasts may be difficult to find
­affected tissues in some infections. Special stains may
assist organism detection.

Laboratory Abnormalities cylindruria are present. Rarely, B. dermatitidis yeasts are identi-
Complete Blood Count fied in the sediment.
The hemogram of dogs or cats with blastomycosis can be unre-
markable. However, a mild, normocytic, normochromic nonre- Cerebrospinal Fluid Analysis
generative anemia is present in many affected animals. Mild to CSF analysis in dogs with CNS blastomycosis can reveal
moderate neutrophilia is also present in many dogs and may be increased total nucleated cell counts and increased CSF protein
accompanied by a mild to moderate bandemia.10,38 Mild mono- concentration.39 Nucleated cells typically consist of a mixture of
cytosis, lymphocytosis, or lymphopenia may be detected. small and large mononuclear cells and neutrophils.

Serum Biochemical Tests Diagnostic Imaging


Serum biochemistry findings in animals with blastomycosis Plain Radiography
include mild to moderate hyperglobulinemia due to a poly- Radiographic patterns in dogs with blastomycosis vary con-
clonal gammopathy, hypoalbuminemia, and, uncommonly, siderably and include unstructured, miliary, or nodular inter-
mild hypercalcemia.10,38 Hypoalbuminemia was present in 70 stitial patterns; a bronchointerstitial pattern; alveolar or mixed
of 91 (77%) of dogs in one study, hyperglobulinemia in 58%, alveolar-interstitial patterns; pulmonary mass lesions (>3 cm in
and hypercalcemia in 14% of dogs.10 diameter); or single or multiple large (0.5 to 2.9 cm) pulmo-
nary nodules that can resemble metastatic pulmonary neopla-
Urinalysis sia (Figure 60-6).10,40,41 Tracheobronchial lymphadenopathy
The urinalysis is usually unremarkable in animals with blas- is present in approximately 25% of dogs.40 Pleural thickening
tomycosis, but occasionally proteinuria, pyuria, hematuria, or and mild pleural effusion are uncommonly identified (≤10% of
CHAPTER 60  Blastomycosis 579

in a cat.42 Intranasal or retrobulbar mass lesions can invade


the cribriform plate or other parts of the skull, with associ-
ated osteolysis. Meningeal or periventricular contrast enhance-
ment may also be present.25,26 MRI findings have not been
extensively described. One dog had a retrobulbar mass lesion
that extended through the calvarium; the lesion was iso- to
hypointense on T2-weighted images, slightly hypointense on
T1-weighted images, and showed strong homogenous contrast
enhancement.29

Microbiologic Tests
Cytologic Examination
Cytologic examination of fine-needle aspirates, respiratory
wash specimens, or body fluids frequently reveals large numbers
of B. dermatitidis yeasts. The yeasts are 8 to 15 µm in diam-
eter, have a thick, refractile cell wall, and exhibit broad-based
budding (Figure 60-8). Daughter cells are nearly as large as the
parent cell when they detach. A pyogranulomatous inflamma-
A
tory response is usually present. Occasionally, organisms are
not seen; the sensitivity of transtracheal lavage for diagnosis of
pulmonary blastomycosis in two studies of 17 and 39 dogs was
76% and 69%,38,41 whereas the sensitivity of fine-needle aspira-
tion of the lung was 46 of 57 (81%).38

Serologic Diagnosis
Serologic assays that detect antibody and those that detect
B. dermatitidis antigen are available on a commercial basis in
North America. The most widely available antibody assay uses
gel immunodiffusion (ID) (see Chapter 2), which detects anti-
bodies to the B. dermatitidis A antigen. Unfortunately, gel ID
assays have variable and generally unacceptable sensitivity for
diagnosis of blastomycosis in dogs and cats, which has ranged
from 17% to 91%.10,31,38,43-45 An enzyme immunoassay that
used crude mold-phase B. dermatitidis as the antigen had a sen-
sitivity of 76.1%,43 and a radioimmunoassay designed to detect
B antibody responses to the BAD-1 antigen had a sensitivity of
92%.45 The specificity of the gel ID assay exceeds 95% based
FIGURE 60-6  Radiographic patterns in pulmonary blastomycosis. A, Lateral thoracic
radiograph from a 2-year-old female spayed Labrador retriever with a 1-week history of on limited studies of healthy dogs or dogs with diagnoses other
cough and inappetence. A diffuse miliary nodular interstitial pattern is present. B, Lateral than blastomycosis.44,45 However, because false-positive test
thoracic radiograph from a 5-year-old female spayed German shepherd dog with leth- results have the potential to occur as a result of exposure and
argy and inappetence. An alveolar pattern is present in the caudal lung lobes, along with recovery in endemic areas or as a result of cross-reactivity to
marked hilar lymphadenopathy. There is also a nodular lesion in the cranial lung lobes just other infectious agents, diagnosis based on positive antibody
ventral to the trachea. (Courtesy Daniel Cronk, University of Minnesota.) serology alone is not recommended.
An assay that detects Blastomyces cell wall galactomannan
antigen (MiraVista Diagnostics, Indianapolis, IN) has largely
affected dogs). Focal bronchiectasis has also been described. In replaced antibody assays for serologic diagnosis of canine blas-
one study, only 2 of 125 dogs with blastomycosis had no ­visible tomycosis. The performance of this assay was evaluated in 46
abnormalities on thoracic radiography. Findings in affected cats dogs with confirmed blastomycosis.43 When urine was used, the
include diffuse miliary or nodular interstitial patterns, lobar sensitivity was 93.5%, whereas when serum was assayed, sensi-
consolidation, and/or pleural effusion.13,31 tivity was 87%. Only 1 of 43 control dogs without blastomyco-
Radiographs of affected bone typically show osteolysis, often sis had a positive test result. In human patients, the sensitivity of
accompanied by periosteal proliferation and soft tissue swelling. urine galactomannan antigen testing was 90%, and specificity
Pathologic fractures may be identified (Figure 60-7). Evidence was 99% in patients without fungal infections. However, 96%
of hypertrophic osteopathy was present bilaterally along the of humans with histoplasmosis tested positive as a result of
humerus, radius, ulna, metacarpi, and phalanges of a dog with cross-reactivity.46 Positive test results can also occur in dogs and
a mass lesion in the right middle lung lobe.22 cats that have histoplasmosis, although the extent to which this
occurs requires further study. It is possible that cross-reactivity
Advanced Imaging may also occur when other mycoses are present.
Computed tomographic findings in dogs with CNS blastomy-
cosis include intra-axial, intranasal, or retrobulbar mass lesions Fungal Culture
that are uniformly or heterogeneously contrast enhancing.25 An B. dermatitidis can be isolated from clinical specimens on rou-
intra-axial contrast-enhancing mass lesion was also described tine fungal media in the laboratory. Growth of a white mold
580 SECTION 3  Fungal and Algal Diseases

A B
FIGURE 60-7  Lateral (A) and anteropalmar (B) radiograph of the right carpus showing an osteolytic and osteoproductive (small arrow) lesion with a pathologic fracture (arrowhead)
and associated soft tissue swelling in the distal radius of a 6-year-old male neutered Labrador retriever with disseminated blastomycosis. (Image courtesy Daniel Cronk, University of
Minnesota.)

the possibility of a dimorphic fungal infection, so that appropriate


precautions are taken.

Molecular Diagnosis Using the Polymerase Chain Reaction


Real-time PCR assays have been developed that rapidly detect
B. dermatitidis in clinical specimens from humans,47,48 but are
not currently used routinely for diagnosis in dogs and cats. The
sensitivity of one assay was 86% when compared with culture
of clinical specimens.48 PCR assays have not been widely applied
to the diagnosis of blastomycosis in dogs and cats.

Pathologic Findings
Gross pathologic findings in dogs with blastomycosis include
thin body condition; characteristic skin lesions; bony prolifera-
tions that may be accompanied by pathologic fracture; enlarge-
ment of the peripheral and/or tracheobronchial lymph nodes;
ocular lesions; lung consolidation; and firm, pale, pulmonary
FIGURE 60-8  Cytology of a fine-needle aspirate of a skin lesion from a dog with nodules or masses that range in size from 1 mm to several centi-
blastomycosis. Blastomyces yeasts (arrows) have a thick wall and exhibit broad-based bud- meters in diameter. Masses may be caseous on cut surface. Ani-
ding. A pyogranulomatous inflammatory response is also present. (Image courtesy Dr. Jed mals with CNS involvement may have masses within the brain,
Overmann, University of Minnesota.) secondary hydrocephalus, or retrobulbar or caudal nasal cavity
granulomas that may invade the cribriform plate and extend
along optic nerves.27,39 Rarely, pleural and/or peritoneal effu-
sion and nodular lesions within abdominal viscera have been
typically appears after incubation for 1 to 3 weeks, but occa- reported.20 Testicular and prostatic masses can also be found.27
sionally incubation periods of up to 5 weeks are required. The Histopathology reveals granulomatous or pyogranulomatous
organism is identified based on the morphology of its conidia, inflammatory infiltrates in a variety of organs, often with intra-
which are round to oval and attached to hyphae (see Figure lesional budding yeasts (Figure 60-9). Multinucleated giant
60-1). Because B. dermatitidis grows as a mycelium in the labo- cells, fibroblasts, and large numbers of lymphocytes may also
ratory, culture is a laboratory health hazard and should be per- be present. Ocular lesions include uveitis, choroiditis, retinal
formed only if necessary. The laboratory should be warned of detachment, retinal degeneration, lens rupture, cataracts, optic
CHAPTER 60  Blastomycosis 581

A B
FIGURE 60-9  Histopathologic findings in dogs with blastomycosis. A, Histopathology of the lymph node from a 10-year-old female spayed Australian shepherd dog with
blastomycosis. Severe pyogranulomatous lymphadenitis is present with moderate numbers of large yeast organisms. (Image courtesy Dr. Catherine Benson, University of Minnesota.)
B, Histopathology of the lung of another Australian shepherd dog that had a pulmonary and a mediastinal abscess due to Blastomyces dermatitidis. Abundant yeast organisms are
present. 1000x oil magnification. (Image courtesy Dr. Patricia Pesavento, University of California, Davis.)

neuritis, and vitritis.27,34 Secondary hepatic and renal amyloido- posaconazole.54,55 Because of its ability to penetrate the CNS
sis was described in one dog.49 and eye, voriconazole has been recommended for treatment of
Organisms are not always detected in lesions, even in human CNS blastomycosis after initial treatment with lipid-
untreated dogs. Special stains such as silver stains, periodic complexed amphotericin B.55
acid–Schiff, and immunohistochemical stains can assist in the Urine Blastomyces galactomannan antigen concentra-
identification of B. dermatitidis yeasts within sections,34,49,50 tion declines with effective antifungal drug treatment.43
but failure to detect yeasts does not rule out blastomycosis. It is not yet clear whether treatment should be continued
until antigenuria is undetectable when radiographic lesions
Treatment and Prognosis resolve earlier. It is possible that the immune system of some
dogs may continue to clear antigen even after treatment is
Antimicrobial Treatment discontinued.
The most widely used treatment for blastomycosis in dogs
is itraconazole, which is effective as a single agent in many Supportive Care
dogs.38,51 Itraconazole has largely replaced the use of ketocon- Other treatments that may be required for treatment of dogs
azole for treatment of blastomycosis. The recommended dose or cats with severe pulmonary blastomycosis are supplemental
of itraconazole for dogs is 5 mg/kg PO q24h. This dose has an oxygen, nebulization and coupage, intravenous fluid therapy,
equivalent efficacy as a dose of 5 mg/kg PO q12h and a lower and, in some cases, mechanical ventilation.38 Mechanical ven-
rate of adverse effects such as anorexia.51 The duration of azole tilation was used to treat 5 (4%) of 125 dogs with pulmonary
treatment should be based on serial monitoring of clinical blastomycosis in one report, none of which survived.38 Dogs
signs and radiographic lesions (e.g., every 4 to 8 weeks). Most with CNS involvement may require treatment with anticon-
dogs require at least 3 to 6 months of treatment, and some vulsants in order to control seizure activity. The concurrent
dogs require treatment for more than 1 year, especially those use of systemic glucocorticoids should also be considered to
with osteoarticular infections or widespread dissemination. control brain inflammation and edema, although whether
Because fluconazole is less active, its use is not recommended this ultimately improves outcome is not known. For animals
for treatment of human blastomycosis, unless itraconazole is that lack CNS involvement, NSAIDs can be used to control
not tolerated.52 Treatment with deoxycholate amphotericin B pyrexia. Topical anti-inflammatory and antiglaucoma agents
or lipid-complexed amphotericin B is also effective and could may be indicated if ocular involvement is present. Enucle-
be considered for dogs with severe disease with widespread ation may ultimately be required if endophthalmitis is present
dissemination, for animals that do not respond to azole mono to control ocular pain and eliminate infection. Treatment of
therapy, or for those that do not tolerate itraconazole (see osteomyelitis or large pulmonary granulomas that are refrac-
Box 9-1).38,53 Cats with blastomycosis have also been treated tory to antifungal chemotherapy may require surgical treat-
with variable success with combinations of amphotericin B ment by amputation or lung lobectomy, respectively.
and azoles or amphotericin B alone.13,31 In human patients,
the use of deoxycholate or lipid-complexed amphotericin B is Prognosis
recommended for severe pulmonary or disseminated disease, Cure rates of 50% to 75% have been reported in dogs with
followed by step-down itraconazole therapy after there has blastomycosis.51,56 An additional 20% of dogs experience
been a satisfactory clinical response.52 Whether protocols that relapse of disease after treatment is discontinued.51 Clinical
include amphotericin B offer a survival advantage over azole signs and radiographic lesions can worsen in the first few days
monotherapy for dogs or cats has not been well studied in a of treatment in some affected dogs,40 possibly as a result of the
prospective fashion. Other drugs that have been used success- inflammatory response to dying organisms. The median time
fully to treat human blastomycosis include voriconazole and for resolution of primary radiographic patterns in dogs with
582 SECTION 3  Fungal and Algal Diseases

A B
FIGURE 60-10  Lateral thoracic radiographs from a young adult male neutered golden retriever with blastomycosis. A, A nodular soft tissue pattern is present diffusely throughout all
lung lobes, which coalesces to alveolar infiltrates in the left caudal lung lobe and right cranial lung lobe. B, Residual pulmonary infiltrates are present in the right cranial lung lobe after 14
months of itraconazole treatment. There is also a large bulla in the left caudal lung lobe (arrows).

pulmonary blastomycosis in one study was 186 days (range, 4 Prevention


to >355 days).40 Significantly longer mean treatment durations
were required for radiographic improvement of large pulmo- Avoidance of specific foci of hyperendemicity where other dogs
nary masses when compared with alveolar and interstitial pat- or humans have contracted blastomycosis may prevent blasto-
terns. The most common radiographic sequela to pulmonary mycosis, but this is not always possible.
blastomycosis is development of one or more pulmonary bullae.
Unstructured interstitial patterns may also persist, presumably Public Health Aspects
as a result of pulmonary fibrosis (Figure 60-10).40
Involvement of the CNS, severe lung disease, and a high band Like dogs, humans acquire B. dermatitidis infection from the
neutrophil count are negative prognostic factors.38,51,56 In one environment. Dogs are considered sentinels for human exposure
study, the median band neutrophil count in dogs that did not and infection,59 although whether strains that infect dogs also
survive was 1200 cells/µL (range, 0 to 3980 cells/µL), whereas infect humans requires further study. The incidence of blasto-
that in survivors was 0 cells/µL (range, 0 to 2380 cells/µL).38 mycosis in dogs is approximately 8 times that in humans, and
In that study, 63% of 125 dogs with blastomycosis survived. disease has been reported in humans and dogs that reside in the
The prognosis for survival in animals with neurologic involve- same household.59,60 Approximately 50% of human infections
ment is particularly poor. The prognosis for resolution of endo- are asymptomatic.52 When illness develops, it typically occurs
phthalmitis is also poor; only 20% of eyes with endophthalmitis 30 to 45 days after exposure and is often a mild, self-limiting
respond favorably to antifungal drug treatment.33 Nevertheless, influenza-like illness with fever and cough. Blastomycosis is sus-
eyes that do not undergo enucleation can ultimately progress to pected in endemic areas when respiratory signs persist and fail
phthisis bulbi after completion of treatment, without recurrence to respond to treatment with antibacterial drugs. Radiographic
of systemic infection.33 abnormalities resemble those in dogs, although hilar lymphade-
nopathy is not often seen. Chronic pulmonary blastomycosis can
Immunity and Vaccination resemble pulmonary neoplasia or tuberculosis. Rarely, blasto-
mycosis-associated acute respiratory distress syndrome (ARDS)
Immunity to blastomycosis is initially dependent on phagocyte develops, which is associated with mortality rates that exceed
function, especially neutrophils and alveolar macrophages, 50%.52 Dissemination to extrapulmonary sites similar to those
which can clear conidia. However, once the organisms have tran- involved in dogs occurs in 20% to 25% of affected humans.
sitioned to the yeast form, control of infection also depends on Serious infections can occur in immunocompromised patients
T lymphocytes, which stimulate macrophages to kill the yeasts.52 such as those with AIDS, solid organ transplant recipients, and
Humoral immunity is not essential for resolution of infec- humans treated with TNF-α blockers (used to treat inflamma-
tion. An experimental vaccine that includes a genetically engi- tory diseases such as Crohn’s disease, psoriasis, and rheumatoid
neered, live-attenuated BAD-1 deletion mutant B. dermatitidis arthritis).52,61,62
strain protected mice from experimental infection, was safe in Inoculation blastomycosis that involves the skin and under-
25 beagles and 78 foxhounds in a field trial, and induced specific lying bone has been described in veterinary personnel after
immune responses to B. dermatitidis antigens.57,58 Although fur- sharps injuries or bite wounds from dogs with blastomycosis.
ther study is required, this vaccine holds promise for prevention One involved a needle-stick injury after a lung aspirate in a dog
of blastomycosis in dogs that reside in hyperendemic regions of with suspected blastomycosis, and another was an accident that
the United States. occurred during a necropsy.63,64 Bite wounds from dogs with
CHAPTER 60  Blastomycosis 583

blastomycosis can result in localized disease; disseminated dis- isolation of microorganisms from dogs and cats may reduce the
ease occurred in a renal transplant recipient.65-67 A veterinary risk of inoculation or laboratory-acquired blastomycosis in vet-
technician developed pulmonary blastomycosis after B. derma- erinary staff. The bandaging of skin lesions has been discouraged
titidis was cultured unexpectedly in a veterinary clinic in-house because it has the potential to promote transition of the organism
laboratory.68 Although these routes of transmission are rare, to the mold form, but this has not been documented to occur.
prevention of sharps injuries, avoidance and proper management The body of deceased pets with blastomycosis should be disposed
of bite wounds, and use of accredited veterinary laboratories for of promptly and by cremation.

CASE EXAMPLE Respiratory: Increased respiratory effort was present. Lung


sounds were increased diffusely on thoracic auscultation
and partially obscured the heart sounds. Markedly
Signalment: “Sam,” a 3-year-old male neutered golden increased respiratory effort was evident when the dog’s
retriever from Burlingame in northern California gait was assessed as part of an orthopedic examination. An
History: Sam was evaluated by the Veterinary Emergency and intermittent nonproductive cough was also appreciated.
Critical Care Service at the University of California, Davis, for All Other Systems: No clinically significant abnormalities were
pulmonary blastomycosis, which had been diagnosed at a detected. No obvious neurologic deficits were noted, but a full
local veterinary clinic. Sam had a 1-week history of lethargy neurologic examination was not performed.
and inappetence. The day after illness was noted, Sam Laboratory Findings: Blood oxygen saturation (pulse
was taken to a local veterinary clinic. Routine blood work oximetry) = 91%, improved to 95% with flow-by
showed only anemia (hematocrit 34%), thrombocytosis supplemental oxygen
(554,000 platelets/µL), and a mildly increased serum alkaline CBC:
phosphatase activity (199 U/L). Thoracic radiographs showed HCT 37.3% (40%-55%)
hilar lymphadenopathy, a mild diffuse miliary interstitial MCV 71.2 fL (65-75 fL)
pattern, and focal alveolar infiltrates in the right cranial and MCHC 35.1 g/dL (33-36 g/dL)
left caudal lung lobe. Cytologic examination of a transtracheal WBC 14,640 cells/µL (6000-13,000 cells/µL)
lavage specimen showed pyogranulomatous inflammation Neutrophils 11,273 cells/µL (3000-10,500 cells/µL)
with moderate numbers of yeasts that had morphology Band neutrophils 586 cells/µL
consistent with Blastomyces dermatitidis. Serology (gel Lymphocytes 1318 cells/µL (1000-4000 cells/µL)
immunodiffusion) for detection of antibodies to B. dermatitidis Monocytes 1318 cells/µL (150-1200 cells/µL)
was negative. Treatment with itraconazole (5 mg/kg PO q24h) Eosinophils 146 cells/µL (0-1500 cells/µL)
was commenced, and after 2 days the dose was increased to Platelets 387,000 platelets/µL (150,000-400,000 platelets/µL)
10 mg/kg PO q24h because of lack of clinical improvement. Rare slight toxicity of band neutrophils was reported.
Subsequently the dog’s rectal temperature increased to 105°F Serum Chemistry Profile:
(40.6°C) and a day later, radiographs showed an increase in the Sodium 148 mmol/L (145-154 mmol/L)
severity of the alveolar infiltrates. Treatment with carprofen Potassium 5.0 mmol/L (4.1-5.3 mmol/L)
(2.4 mg/kg PO q12h) was initiated, and subcutaneous fluids Chloride 116 mmol/L (105-116 mmol/L)
were administered. This was associated with resolution Bicarbonate 15 mmol/L (16-26 mmol/L)
of pyrexia, but the dog remained lethargic and exercise Phosphorus 6.6 mg/dL (3.0-6.2 mg/dL)
intolerant and the owners noted right pelvic limb lameness. Calcium 9.9 mg/dL (9.9-11.4 mg/dL)
The dog was referred for further evaluation. BUN 11 mg/dL (8-21 mg/dL)
Approximately 6 weeks before he became ill, Sam had been Creatinine 0.5 mg/dL (0.5-1.6 mg/dL)
taken to Georgian Bay in Ontario, Canada, for 1 week. He Glucose 81 mg/dL (60-104 mg/dL)
had also been in the Vermont region for 2 months before Total protein 5.6 g/dL (5.4-7.4 g/dL)
that time. Other travel had been limited to various bayside Albumin 1.8 g/dL (2.9-4.2 g/dL)
and mountainous parts of northern California. The owner’s Globulin 3.8 g/dL (2.3-4.4 g/dL)
brother’s dog, which had traveled with Sam to Ontario and ALT 15 U/L (19-67 U/L)
Vermont, had also been diagnosed with blastomycosis 3 AST 32 U/L (21-54 U/L)
weeks earlier. ALP 148 U/L (15-127 U/L)
Physical Examination: GGT 0 U/L (0-6 U/L)
Body Weight: 41.5 kg. Cholesterol 318 mg/dL (135-345 mg/dL)
General: Quiet, alert and responsive. T = 103.5°F (39.7°C), HR = Total bilirubin 0.4 mg/dL (0-0.4 mg/dL).
120 beats/min, panting, CRT <2 s, mucous membranes were Urinalysis: SGr 1.015; pH 5.0, no protein (SSA), 1+ bilirubin,
moist and slightly cyanotic. no hemoprotein, no glucose, no WBC, rare RBC/HPF, rare
Eyes, Ears, Nose, and Throat: No clinically significant transitional epithelial cells, and a few amorphous crystals
abnormalities were detected. A fundoscopic examination were seen.
with dilated pupils was unremarkable.
Musculoskeletal: BCS 4/9; no obvious lameness or pain on
palpation was appreciated.

Continued
584 SECTION 3  Fungal and Algal Diseases

Imaging Findings: At a recheck 1 month after the date of first evaluation, Sam re-
Thoracic Radiographs: A nodular soft tissue pattern was mained somewhat lethargic and exercise intolerant, but had
present diffusely throughout all lung lobes, which coalesced gained 3 kg. The frequency of cough had decreased from
to alveolar infiltrates in the left caudal lung lobe and right approximately 8 times a day to once or twice daily over the
cranial lung lobe (see Figure 60-10). The cardiac silhouette previous 2 weeks. An arterial blood gas showed a pO2 of 93.5
was mildly enlarged. Increased soft tissue opacity was noted mm Hg, hematocrit was 27.8% with 39,400 reticulocytes/µL,
in the perihilar region. and there was persistent hypoalbuminemia (2.0 mg/dL) and
Abdominal Ultrasound: The spleen was mildly enlarged, and hyperglobulinemia (4.8 mg/dL). There was mild radiographic
there was mild mesenteric lymphadenopathy. improvement in the pulmonary lesions. Treatment with itra-
Diagnosis: Acute, severe pulmonary blastomycosis. conazole was continued for an additional 14 months, with
Treatment: Sam was placed in an oxygen cage with an inspired serial monitoring of thoracic radiographs at 1, 2, 3, 8, and 14
oxygen concentration (FiO2) of 40% to 60%. An arterial blood months. At the 14-month time point, Sam had returned to his
gas revealed a pH of 7.301, pCO2 of 36.6 mm Hg, pO2 of normal energetic self, weighed 45 kg, but had persistent mild
79.1 mm Hg, HCO3 of 16.8 mmol/L, base deficit of 7.8 mmol/L, infiltrates in the right cranial lung lobe on thoracic radiographs
and measured O2 saturation of 92.8%. The dog was also and a bulla in the left cranial lung lobe. There was also a mild to
treated with crystalloid fluids (lactated Ringer’s solution with moderate generalized interstitial pattern with multiple small
20 mEq/L KCl; 80 mL/hr, IV) and nebulization and coupage soft tissue opaque nodules that had not changed from the
(q6h). For the first 24 hours, respiratory rate ranged from 90 previous evaluation. One month after the itraconazole was
to 105 breaths/min and a nonproductive cough occurred discontinued, thoracic radiographs remained unchanged. Re-
every few hours. The dog drank water, but was inappetent. lapse did not occur. Sam was ultimately euthanized at 8 years
Treatment with lipid-complexed amphotericin B was of age as a result of an unrelated neoplastic disease.
initiated (1 mg/kg IV on a Monday-Wednesday-Friday basis) Comments: This case is an example of acute pulmonary
and carprofen administration continued. By day 4, Sam was blastomycosis that followed travel to an endemic area.
afebrile, eating, and oxygen saturation was 96% with an FiO2 Another dog that traveled to the same area also developed
of 46%. However, increased respiratory effort developed disease. Sam appeared to require more aggressive
when oxygen supplementation was discontinued, and therapy than itraconazole alone, so amphotericin B was
arterial blood gas analysis showed a pH of 7.335, pCO2 of 27.2 instituted, which was followed by gradual recovery from
mm Hg, pO2 of 65.8 mm Hg, HCO3 of 13.9 mmol/L, and base the infection. Although lipid-complexed amphotericin B
deficit of 10.6 mmol/L (room air). By day 5, Sam was playing was used, treatment with deoxycholate amphotericin B
with toys in the oxygen cage, and his appetite was excellent. may also have led to cure. No evidence of dissemination
Treatment with itraconazole (5 mg/kg PO q24h) was resumed was clearly identified. The lameness observed by the client
in addition to the amphotericin B. By day 8, oxygen saturation may have represented weakness or exercise intolerance,
was 95% with an FiO2 of 30%. Thoracic radiographs showed but it is possible that bone involvement was present but
persistent radiographic abnormalities, organization of the not evident radiographically. The case also illustrates
alveolar infiltrates, and apparent bronchiectasis. Survey the residual pulmonary lesions that can develop in dogs
radiographs of the appendicular skeleton showed no with pulmonary blastomycosis, and the need for serial
evidence of osteomyelitis. The dog was discharged after 11 radiographic evaluations to assess these lesions. Although
days of hospitalization (cumulative amphotericin B dose of not performed in this dog, serial monitoring of antigenuria
6 mg/kg). An additional seven treatments of amphotericin may also ultimately help determine the most appropriate
B were then administered at the local veterinary clinic. Serial time to discontinue therapy.
renal panels showed no evidence of nephrotoxicity.

SUGGESTED READINGS
4. Iyer PKR. Pulmonary blastomycosis in a dog in India. Indian J Vet
Arceneaux KA, Taboada J, Hosgood G. Blastomycosis in dogs: 115 Pathol. 1983;7:60-62.
cases (1980-1995). J Am Vet Med Assoc. 1998;213:658-664. 5. Kucher I, Christiansen E, Schneider-Haiss M. Blastomykose bei
Crews LJ, Feeney DA, Jessen CR, et al. Radiographic findings in dogs einer Rottweiler-Hündin. Kleintierpraxis. 1998;43:627-631.
with pulmonary blastomycosis: 125 cases (1989-2006). J Am Vet 6. Meece JK, Anderson JL, Klein BS, et al. Genetic diversity in Blasto-
Med Assoc. 2008;232:215-221. myces dermatitidis: implications for PCR detection in clinical and
Smith JA, Kauffman CA. Blastomycosis. Proc Am Thorac Soc. 2010; environmental samples. Med Mycol. 2010;48:285-290.
7:173-180. 7. Meece JK, Anderson JL, Fisher MC, et al. Population genetic struc-
Spector D, Legendre AM, Wheat J, et  al. Antigen and antibody test- ture of clinical and environmental isolates of Blastomyces der-
ing for the diagnosis of blastomycosis in dogs. J Vet Intern Med. matitidis, based on 27 polymorphic microsatellite markers. Appl
2008;22:839-843. Environ Microbiol. 2011;77:5123-5131.
8. Meece JK, Anderson JL, Gruszka S, et al. Variation in clinical phe-
REFERENCES notype of human infection among genetic groups of Blastomyces
1. Gilchrist TC. Protozoan dermatitis. J Cutan Gen Dis. 1894; dermatitidis. J Infect Dis. 2013;207:814-822.
12:496-499. 9. Rudmann DG, Coolman BR, Perez CM, et al. Evaluation of risk
2. Harasen GL, Randall JW. Canine blastomycosis in southern Sas- factors for blastomycosis in dogs: 857 cases (1980-1990). J Am Vet
katchewan. Can Vet J. 1986;27:375-378. Med Assoc. 1992;201:1754-1759.
3. Hoff B. North American blastomycosis in two dogs in Saskatch- 10. Arceneaux KA, Taboada J, Hosgood G. Blastomycosis in dogs: 115
ewan. Can Vet J. 1973;14:122-123. cases (1980-1995). J Am Vet Med Assoc. 1998;213:658-664.
CHAPTER 60  Blastomycosis 585

11. Baumgardner DJ, Paretsky DP. The in vitro isolation of Blastomy- 36. McEwen SA, Hulland TJ. Cerebral blastomycosis in a cat. Can Vet
ces dermatitidis from a woodpile in north central Wisconsin, USA. J. 1984;25:411-413.
Med Mycol. 1999;37:163-168. 37. Nasisse MP, van Ee RT, Wright B. Ocular changes in a cat with dis-
12. Blondin N, Baumgardner DJ, Moore GE, et  al. Blastomycosis in seminated blastomycosis. J Am Vet Med Assoc. 1985;187:629-631.
indoor cats: suburban Chicago, Illinois, USA. Mycopathologia. 38. Crews LJ, Feeney DA, Jessen CR, et al. Utility of diagnostic tests for
2007;163:59-66. and medical treatment of pulmonary blastomycosis in dogs: 125
13. Breider MA, Walker TL, Legendre AM, et al. Blastomycosis in cats: cases (1989-2006). J Am Vet Med Assoc. 2008;232:222-227.
five cases (1979-1986). J Am Vet Med Assoc. 1988;193:570-572. 39. Nafe LA, Turk JR, Carter JD. Central nervous system involvement of
14. Davies C, Troy GC. Deep mycotic infections in cats. J Am Anim blastomycosis in the dog. J Am Anim Hosp Assoc. 1983;19:933-936.
Hosp Assoc. 1996;32:380-391. 40. Crews LJ, Feeney DA, Jessen CR, et  al. Radiographic findings in
15. Gray NA, Baddour LM. Cutaneous inoculation blastomycosis. dogs with pulmonary blastomycosis: 125 cases (1989-2006). J Am
Clin Infect Dis. 2002;34:E44-E49. Vet Med Assoc. 2008;232:215-221.
16. Marcellin-Little DJ, Sellon RK, Kyles AE, et al. Chronic localized 41. McMillan CJ, Taylor SM. Transtracheal aspiration in the diagno-
osteomyelitis caused by atypical infection with Blastomyces derma- sis of pulmonary blastomycosis (17 cases: 2000-2005). Can Vet J.
titidis in a dog. J Am Vet Med Assoc. 1996;209:1877-1879. 2008;49:53-55.
17. Finkel-Jimenez B, Wuthrich M, Klein BS. BAD1, an essential vir- 42. Smith JR, Legendre AM, Thomas WB, et  al. Cerebral Blas-
ulence factor of Blastomyces dermatitidis, suppresses host TNF- tomyces dermatitidis infection in a cat. J Am Vet Med Assoc.
alpha production through TGF-beta-dependent and -independent 2007;231:1210-1214.
mechanisms. J Immunol. 2002;168:5746-5755. 43. Spector D, Legendre AM, Wheat J, et al. Antigen and antibody test-
18. Nosanchuk JD, van Duin D, Mandal P, et  al. Blastomyces der- ing for the diagnosis of blastomycosis in dogs. J Vet Intern Med.
matitidis produces melanin in  vitro and during infection. FEMS 2008;22:839-843.
Microbiol Lett. 2004;239:187-193. 44. Legendre AM, Becker PU. Evaluation of the agar-gel immunodif-
19. Wuthrich M, Finkel-Jimenez B, Brandhorst TT, et al. Analysis of fusion test in the diagnosis of canine blastomycosis. Am J Vet Res.
non-adhesive pathogenic mechanisms of BAD1 on Blastomyces 1980;41:2109-2111.
dermatitidis. Med Mycol. 2006;44:41-49. 45. Klein BS, Squires RA, Lloyd JK, et  al. Canine antibody response
20. Nielsen C, Olver CS, Schutten MM, et  al. Diagnostic peritoneal to Blastomyces dermatitidis WI-1 antigen. Am J Vet Res.
lavage for identification of blastomycosis in a dog with peritoneal 2000;61:554-558.
involvement. J Am Vet Med Assoc. 2003;223:1623-1627:1600. 46. Connolly P, Hage CA, Bariola JR, et al. Blastomyces dermatitidis
21. Menges RW, Furcolow ML, Selby LA, et al. Clinical and epidemio- antigen detection by quantitative enzyme immunoassay. Clin Vac-
logic studies on seventy-nine canine blastomycosis cases in Arkan- cine Immunol. 2012;19:53-56.
sas. Am J Epidemiol. 1965;81:164-179. 47. Sidamonidze K, Peck MK, Perez M, et al. Real-time PCR assay for
22. Brockus CW, Hathcock JT. Hypertrophic osteopathy associated identification of Blastomyces dermatitidis in culture and in tissue. J
with pulmonary blastomycosis in a dog. Vet Radiol Ultrasound. Clin Microbiol. 2012;50:1783-1786.
1988;29:184-188. 48. Babady NE, Buckwalter SP, Hall L, et al. Detection of Blastomy-
23. Totten AK, Ridgway MD, Sauberli DS. Blastomyces dermatitidis ces dermatitidis and Histoplasma capsulatum from culture isolates
prostatic and testicular infection in eight dogs (1992-2005). J Am and clinical specimens by use of real-time PCR. J Clin Microbiol.
Anim Hosp Assoc. 2011;47:413-418. 2011;49:3204-3208.
24. Ditmyer H, Craig L. Mycotic mastitis in three dogs due to Blasto- 49. Sherwood BF, LeMay JC, Castellanos RA. Blastomycosis
myces dermatitidis. J Am Anim Hosp Assoc. 2011;47:356-358. with secondary amyloidosis in the dog. J Am Vet Med Assoc.
25. Hecht S, Adams WH, Smith JR, et al. Clinical and imaging findings 1967;150:1377-1381.
in five dogs with intracranial blastomycosis (Blastomyces derma- 50. Sekhon AS, Marien GR, Easton B, et  al. A canine case of
titidis). J Am Anim Hosp Assoc. 2011;47:241-249. North American blastomycosis in Alberta, Canada. Mycoses.
26. Saito M, Sharp NJ, Munana K, et al. CT findings of intracranial 1988;31:454-458.
blastomycosis in a dog. Vet Rad Ultrasound. 2002;43:16-21. 51. Legendre AM, Rohrbach BW, Toal RL, et  al. Treatment of
27. Wilson RW, van Dreumel AA, Henry JN. Urogenital and ocular blastomycosis with itraconazole in 112 dogs. J Vet Intern Med.
lesions in canine blastomycosis. Vet Pathol. 1973;10:1-11. 1996;10:365-371.
28. Salinardi BJ, Marretta SM, McCullough SM, et  al. Pharyngeal- 52. Smith JA, Kauffman CA. Blastomycosis. Proc Am Thorac Soc.
laryngeal blastomycosis in a dog. J Vet Dent. 2003;20:146-147. 2010;7:173-180.
29. Baron ML, Hecht S, Westermeyer HD, et al. Intracranial extension 53. Krawiec DR, McKiernan BC, Twardock AR, et  al. Use of an
of retrobulbar blastomycosis (Blastomyces dermatitidis) in a dog. amphotericin B lipid complex for treatment of blastomycosis in
Vet Ophthalmol. 2011;14:137-141. dogs. J Am Vet Med Assoc. 1996;209:2073-2075.
30. McGuire NC, Vitsky A, Daly CM, et al. Pulmonary thromboembo- 54. Proia LA, Harnisch DO. Successful use of posaconazole for
lism associated with Blastomyces dermatitidis in a dog. J Am Anim treatment of blastomycosis. Antimicrob Agents Chemother.
Hosp Assoc. 2002;38:425-430. 2012;56:4029.
31. Miller PE, Miller LM, Schoster JV. Feline blastomycosis: a report 55. Bariola JR, Perry P, Pappas PG, et al. Blastomycosis of the central
of three cases and literature review (1961 to 1988). J Am Anim nervous system: a multicenter review of diagnosis and treatment in
Hosp Assoc. 1990;26:417-424. the modern era. Clin Infect Dis. 2010;50:797-804.
32. Bromel C, Sykes JE. Epidemiology, diagnosis, and treatment 56. Legendre AM, Selcer BA, Edwards DF, et al. Treatment of canine
of blastomycosis in dogs and cats. Clin Tech Small Anim Pract. blastomycosis with amphotericin B and ketoconazole. J Am Vet Med
2005;20:233-239. Assoc. 1984;184:1249-1254.
33. Bloom JD, Hamor RE, Gerding Jr PA. Ocular blastomycosis 57. Wuthrich M, Krajaejun T, Shearn-Bochsler V, et al. Safety, toler-
in dogs: 73 cases, 108 eyes (1985-1993). J Am Vet Med Assoc. ability, and immunogenicity of a recombinant, genetically engi-
1996;209:1271-1274. neered, live-attenuated vaccine against canine blastomycosis. Clin
34. Hendrix DV, Rohrbach BW, Bochsler PN, et al. Comparison of his- Vaccine Immunol. 2011;18:783-789.
tologic lesions of endophthalmitis induced by Blastomyces derma- 58. Wuthrich M, Filutowicz HI, Klein BS. Mutation of the WI-1 gene
titidis in untreated and treated dogs: 36 cases (1986-2001). J Am yields an attenuated Blastomyces dermatitidis strain that induces
Vet Med Assoc. 2004;224:1317-1322. host resistance. J Clin Invest. 2000;106:1381-1389.
35. Meschter C, Heiber K. Blastomycosis in a cat in lower New York 59. Sarosi GA, Eckman MR, Davies SF, et al. Canine blastomycosis as
State. Cornell Vet. 1989;79:259-262. a harbinger of human disease. Ann Intern Med. 1979;91:733-735.
586 SECTION 3  Fungal and Algal Diseases

60. Baumgardner DJ, Paretsky DP. Blastomycosis: more evidence for 65. Gnann Jr JW, Bressler GS, Bodet 3rd CA, et al. Human blastomy-
exposure near one’s domicile. WMJ. 2001;100:43-45. cosis after a dog bite. Ann Intern Med. 1983;98:48-49.
61. Pappas PG. Blastomycosis in the immunocompromised patient. 66. Butka BJ, Bennett SR, Johnson AC. Disseminated inoculation
Semin Respir Infect. 1997;12:243-251. blastomycosis in a renal transplant recipient. Am Rev Respir Dis.
62. Gauthier GM, Safdar N, Klein BS, et  al. Blastomycosis in solid 1984;130:1180-1183.
organ transplant recipients. Transpl Infect Dis. 2007;9:310-317. 67. Jaspers RH. Letter: Transmission of Blastomyces from animals to
63. Ramsey DT. Blastomycosis in a veterinarian. J Am Vet Med Assoc. man. J Am Vet Med Assoc. 1974;164:8.
1994;205:968. 68. Cote E, Barr SC, Allen C. Possible transmission of Blastomy-
64. Graham Jr WR, Callaway JL. Primary inoculation blastomycosis in ces dermatitidis via culture specimen. J Am Vet Med Assoc.
a veterinarian. J Am Acad Dermatol. 1982;7:785-786. 1997;210:479-480.
CHAPTER 61

Histoplasmosis
Jane E. Sykes and Joseph Taboada

and H. capsulatum var. farciminosum—although the phyloge-


Overview of Histoplasmosis netic value of these variants has been debated.6,9 H. capsulatum
First Described: 1905, Panama (by Samuel Darling), in a per- var. duboisii occurs primarily in Africa and Japan. H. capsula-
son from Martinique, where it was initially mistaken for a tum var. farciminosum infections occur primarily in horses, but
protozoal pathogen.1 have also been reported in humans and dogs.
H. capsulatum grows best in soil where temperatures are
Cause: Histoplasma capsulatum (an ascomycete)
between 22°C and 29°C, where annual rainfall is 35 to 50 inches,
Affected Hosts: Many mammalian species including dogs, and where relative humidity is 67% to 87%. These conditions
cats, and humans are typically found between latitudes of 45 degrees north to
Geographic Distribution: Worldwide; but especially the Ohio, 30 degrees south.11 H. capsulatum can be found in the intestinal
Missouri, Tennessee, and Mississippi river valleys of the tracts and guano of bats, which are the primary reservoir of the
United States; and Latin America organism and serve to disseminate it geographically.12 Bat caves
can maintain perfect growth conditions for H. capsulatum.
Mode of Transmission: Inhalation of microconidia from the Although H. capsulatum can be found in high concentrations in
environment decaying avian guano (especially around blackbird or starling
Major Clinical Signs: Cough, tachypnea, organomegaly, gas- roosts and chicken coops), it is not found in fresh feces or shed
trointestinal signs, pallor; signs are often non-specific in in the feces of birds.
cats. Cats are as susceptible, or slightly more susceptible, to histo-
Differential Diagnoses: Other deep mycoses, protothecosis, plasmosis than dogs. Affected cats can be as young as 2 months
mycobacterial infections, hemic neoplasia, leishmaniosis, and, in some cases, older than 15 years.13 The mean age of
histiocytic colitis affected cats has varied from around 4 years to 9 years.14 For
dogs, a mean age of 4.3 years was reported (range, 5 months to
Human Health Significance: H. capsulatum also causes dis- 10 years).15 Dogs in the 2- to 4-year and 4- to 7-year age groups
ease in humans but direct transmission from animals to were more likely to develop histoplasmosis than dogs less than
humans does not occur 2 years of age.16 Sporting or working dogs may be at greater
risk of infection because of increased exposure; in a study using
the Veterinary Medical Database, pointers were strongly over-
represented.16 Weimaraners and Brittany spaniels were also at
Etiology and Epidemiology increased risk. Among cats, Persian cats may be slightly over-
represented. A sex predisposition has not been clearly iden-
Histoplasma capsulatum is a dimorphic, soil-borne fungus that tified in dogs or cats, but more female than male dogs were
is found worldwide, but especially along the Mississippi, Mis- affected in some case series.14,15,17-19 This contrasts to human
souri, Tennessee, and Ohio river valleys of the United States, as histoplasmosis and other systemic mycoses in dogs, which affect
well as in Latin America (Figure 61-1). Endemic areas within males more often than females.20 The overwhelming majority of
the United States span the central portion of the United States cats with histoplasmosis are retrovirus negative, but a signifi-
from western Virginia to central Texas. Disease has also been cant percentage of cats in two studies (16% and 28%, respec-
reported in cats or dogs from northern California2; Brazil3; tively) were co-infected with FeLV.13,14 A small percentage of
Italy4; Queensland in Australia5; Japan6; and western Can- cats have underlying comorbidities such as lymphoma, feline
ada.7,8 Eight clades of the organism have been identified by infectious peritonitis, or a history of glucocorticoid treatment.13
genetic analysis: North American (class 1 and class 2), Latin Comorbidities described in dogs include dirofilariasis15 and
American (group A and group B), and one each of Australian, metastatic carcinoma.6 Disease can occur in cats that are housed
Indonesian, Eurasian, and African. These may vary in virulence. exclusively indoors.
All except the Eurasian clade, which is derived from the Latin
American group A clade, may be distinct phylogenetic species.9 Clinical Features
A possible ninth clade, which is related to, but distinct from,
North American class 1 also appears to infect cats in North Amer- Signs and Their Pathogenesis
ica outside the accepted endemic range.10 Three variants have In the soil, the mycelial phase of H. capsulatum forms macro-
also been described based on pathogenicity and morphology— conidia and microconidia (Figure 61-2). The microconidia are
H. capsulatum var. capsulatum, H. capsulatum var. duboisii, thought to be the form inhaled by mammalian hosts, because

587
588 SECTION 3  Fungal and Algal Diseases

they are small enough (2 to 5 µm) to enter the terminal bronchi- on alveolar macrophages and is phagocytized. It then replicates
oles and alveoli. Within the lungs (i.e., at a temperature of 37°C), within these cells through control of the phagolysosomal envi-
the microconidia transition to a unicellular yeast, which repli- ronment and ultimately destroys them, with subsequent replica-
cates by budding. The fungus binds to CD11-CD18 integrins tion in other resident alveolar macrophages and inflammatory
phagocytes that are recruited to the lung.21 Acquisition of iron
by the fungus is essential for growth in  vivo.22 Some animals
control the initial infection but remain latently infected with
small numbers of yeasts. Subsequent immune suppression can
lead to reactivation of infection years later. Thus, the incubation
period can range from 2 to 3 weeks to as long as several years.
The clinical manifestations and rate of disease progression
depend on the host immune response to infection, the infectious
dose, and the Histoplasma strain, which can vary geographi-
cally. The vast majority of infections are probably subclinical.23
Replication of the organism leads to a granulomatous inflam-
matory response in the lungs. Occasionally, the inflammatory
response is followed by fibrosis and scarring. The yeasts migrate
to local lymph nodes (such as the hilar lymph node) and other
tissues that contain mononuclear cells, such as the liver and
spleen. When the cell-mediated immune response is defective
or absent, disseminated disease occurs (referred to in human
patients as progressive disseminated histoplasmosis [PDH]20),
which may be acute or chronic. In addition to lymph nodes,
liver, and spleen, other common sites of dissemination are the
bone marrow, small and/or large intestinal tract, pancreas, skin,
Highly endemic
bones, central nervous system (CNS), and eyes. Dogs with histo-
Moderately endemic plasmosis in the United States appear to be particularly suscepti-
Suspected endemic
ble to intestinal involvement. Severe infiltration of the small and
large intestines leads to malabsorption, diarrhea, severe weight
loss, and hematochezia, often without evidence of respiratory
involvement.
Clinical signs in cats are commonly vague and nonspecific,
such as weight loss, inappetence, weakness, dehydration, and
fever.13,14 Approximately 40% of cats show respiratory signs
such as dyspnea and tachypnea, and to a lesser extent, cough
FIGURE 61-1  Approximate geographic distribution of histoplasmosis in the and nasal discharge. However, respiratory signs may be absent
Americas.

Lung

Lymph node Blood

FIGURE 61-2  Life cycle of Histoplasma capsulatum. In the environment, the organism forms macroconidia (arrowhead, inset) and microconidia (arrows, inset). The microconidia are
thought to be inhaled into the lungs of mammalian hosts. Within the lung, the organisms transform into yeasts that infect alveolar macrophages and then disseminate to local lymph nodes
and the systemic circulation.
CHAPTER 61  Histoplasmosis 589

in some cats with dissemination of infection to nonpulmonary masses, and/or pain on palpation of the abdomen. Ocular signs
sites. Ocular signs such as chorioretinitis and/or posterior or include chorioretinitis, optic neuritis, anterior uveitis, retinal
anterior uveitis occur in approximately one quarter of cats with detachment, panophthalmitis, and glaucoma (Figure 61-4).
histoplasmosis, and around 20% of cats have signs of skeletal In dogs, lethargy, fever, thin body condition, and pale mucous
involvement.13,14,17 Nodular or ulcerated and draining skin membranes are most often found on physical examination. Other
lesions, peripheral lymphadenopathy, vomiting, diarrhea, oral physical examination abnormalities include dehydration, hepa-
ulceration, myelopathy, and/or hematuria due to bladder wall tomegaly, icterus, and/or ascites; tachypnea, increased respira-
involvement have also been reported.4,14,17,24-29 Clinical signs tory effort, and increased lung sounds; lymphadenomegaly that
associated with gastrointestinal involvement are much less likely involves one or more peripheral lymph nodes; abnormal stool
to occur in cats than in dogs. A high proportion of affected cats quality on rectal examination (diarrhea, hematochezia, or melena);
that were necropsied had severe disseminated disease.13 nasal and ocular discharge; tachycardia; tongue lesions; and
In dogs, diarrhea, decreased appetite, weight loss, lethargy, cutaneous nodules, ulceration, or draining tracts.5,8,15,18,30,37,38
fever (up to 40°C or 104°F), and mucosal pallor are the most Ocular examination can reveal uveitis, evidence of optic neuri-
common clinical signs.7,8,15,18 In addition to inappetence, weight tis, or chorioretinitis with retinal detachment.18,39,40 One dog
loss, and diarrhea, other clinical signs referable to the gastro- was seen for lameness, hyperemia, and edema of the left pelvic
intestinal tract include melena, tenesmus, hematochezia, dys- limb because of a sclerosing pyogranulomatous inflammatory
chezia, and/or increased frequency of defecation.8,15,18 Profuse mass in the inguinal region that resulted in venous occlusion.37
diarrhea may be chronic and persist for several months. Hilar Rarely, neurologic signs such as ataxia, obtundation, head tilt,
lymphadenopathy is common, and may be an incidental find-
ing on thoracic radiographs, or it may be severe and contrib-
ute to signs of cough. Other signs include respiratory difficulty,
icterus, vomiting, hepatomegaly, lymphadenomegaly, nasal dis-
charge, ocular signs, polyuria and polydipsia, lameness due to
osteomyelitis, and neurologic signs such as seizures or paralysis/
paresis.8,15,30-32 Affected dogs in Japan have had chronic cutane-
ous or gingival lesions in the absence of pulmonary or gastro-
intestinal involvement,33-36 although disseminated disease has
also been described.6 Skin nodules were also reported in an Aus-
tralian dog with histoplasmosis.5 Varied clinical manifestations
of histoplasmosis in different parts of the world as a result of
strain variation in H. capsulatum also occur in humans.20

Physical Examination Findings


Physical examination findings in cats with histoplasmosis can
be minimal or can include thin body condition or emaciation;
fever; pale and/or rarely icteric mucous membranes; peripheral
lymphadenopathy; respiratory signs such as dyspnea, tachy-
pnea, and increased or decreased lung sounds; single or multiple A
cutaneous nodules, ulcerations, and/or draining tracts (Figure
61-3); lameness; and detection of organomegaly, abdominal

B
FIGURE 61-4  Ocular changes in cats with histoplasmosis. A, Blepharitis. B, Granulo-
matous chorioretinitis with multifocal pigmented lesions. (A, From Ketring KL, Glaze MB.
FIGURE 61-3  Cutaneous lesion on the eyelid of a cat with histoplasmosis. (Courtesy Atlas of Feline Ophthalmology. 2nd ed. Hoboken, NJ: Wiley-Blackwell; 2012. B, Courtesy of
Dr. Amy Grooters, Louisiana State University.) Dr. Mary Belle Glaze, Gulf Coast Animal Eye Clinic, Houston, TX.)
590 SECTION 3  Fungal and Algal Diseases

nystagmus, strabismus, seizures, facial paralysis, and tetraparesis white cell count,14,43 but leukocytosis due to neutrophilia
may be present on physical examination as a result of hepatic and monocytosis may be more common in affected dogs.15,44
encephalopathy or meningoencephalitis.40-42 Increased numbers of band neutrophils with toxic neutrophils,
lymphopenia, monocytopenia, and thrombocytopenia can be
Diagnosis present in some severely affected animals.14,17,18,43,45 Thrombo-
cytopenia can be a result of platelet consumption, sequestration,
Laboratory Abnormalities and/or myelophthisis. Rarely, eosinophilia is present.46
Complete Blood Count
The CBC in dogs and cats with histoplasmosis reflects the pres- Serum Biochemical Tests
ence of systemic inflammation, gastrointestinal hemorrhage, Mild to severe hypoalbuminemia is present in most (>75%)
and/or bone marrow infiltration by fungal organisms. In both affected dogs and cats. Cats with liver involvement may have
dogs and cats, anemia is a common abnormality, may be mild increased activities of serum ALT and AST.17,43 Hyperbilirubi-
to severe, and is usually normocytic, normochromic, and non- nemia and increased serum ALP activity are rarely reported.17
regenerative.14,15,17,18,43,44 Increased numbers of nucleated Hypercalcemia and hyperglobulinemia have been described in
erythrocytes occur in some affected dogs.8,44 The total white a few affected cats.17 Hyperglobulinemia; mild to moderate
cell count or neutrophil count may be normal, increased, or increases in the activity of serum ALT, AST, ALP, and GGT;
decreased.15,17,18,43 Most affected cats have a normal or decreased and hyperbilirubinemia can be found in dogs.

B
FIGURE 61-5  Radiographic abnormalities in feline pulmonary histoplasmosis. A, Lateral thoracic radiograph from an 8-month-old intact male domestic shorthair cat with dis-
seminated histoplasmosis. There is a diffuse bronchointerstitial pulmonary pattern and evidence of sternal lymphadenopathy (arrow). In the viewable abdomen, marked splenomegaly
can be appreciated. B, Lateral thoracic radiograph from a 7-month-old male neutered domestic shorthair cat with histoplasmosis that was evaluated for tachypnea and dyspnea of
10 days duration. There is a diffuse, severe, interstitial-to-coalescing and patchy pulmonary pattern that obscures visualization of the pulmonary vasculature and cardiac contours. (Courtesy
of Lorrie Gaschen, Louisiana State University.)
CHAPTER 61  Histoplasmosis 591

Urinalysis marrow is a common site to yield organisms in cats, even when


Results of urinalysis are usually within normal limits, but low hematologic abnormalities are minimal.
urine specific gravity and bilirubinuria may be present in dogs Cytologically, the yeasts are 2 to 4 µm in diameter, oval,
with hepatic involvement. have a basophilic center, and are surrounded by a clear halo,
which results from shrinkage artifact (see Figure 61-7). They
Coagulation Testing are similar in size to Sporothrix spp., although different in
A few dogs with acute PDH have had coagulation abnormalities shape. A variety of stains can be used, such as Diff-Quik and
characterized by increased PT and PTT and fibrin degradation Wright stains. The organism is not encapsulated, despite its
product or D-dimer concentrations.31,44 Coagulation abnormal- name. Organisms may not be identified within chronic, fibros-
ities in dogs with histoplasmosis may result from disseminated ing lesions.37,49
intravascular coagulation and possibly severe hepatic involve-
ment (see also Canine Case Example). Serologic Testing
Both antibody and antigen (ELISA) assays are available for
Diagnostic Imaging serodiagnosis of histoplasmosis. Antibody assays for Histo-
Plain Radiography plasma capsulatum are based on gel immunodiffusion or com-
In cats, thoracic radiographic findings in histoplasmosis usually plement fixation and have had poor diagnostic sensitivity.13,15,17
consist of diffuse, linear, nodular, or miliary interstitial pat- False positives reportedly occur in recovered animals, although
terns, but mixed interstitial-alveolar-bronchial patterns, pleu- the specificity of antibody assays in dogs and cats is not well
ral effusion, or an absence of abnormal findings can also occur documented. In human patients, acute and convalescent anti-
(Figure 61-5).14,19,43,47 Plain thoracic radiographs in dogs may body testing is useful for diagnosis of acute pulmonary histo-
show alveolar, interstitial, and/or bronchial patterns, tracheo- plasmosis, and a single positive antibody titer is used as an aid
bronchial lymphadenopathy, lung lobe consolidation, and/or for diagnosis of chronic histoplasmosis. In particular, a posi-
rarely pleural effusion.31,45,47,48 Lesions in dogs sometimes min- tive CSF antibody titer can be useful for diagnosis of culture-
eralize, which does not seem to occur in cats. Abdominal radio- negative chronic H. capsulatum meningitis. An ELISA assay for
graphs may show hepatomegaly, splenomegaly, or decreased H. capsulatum antigen (MiraVista Diagnostics, Indianapolis,
serosal detail due to ascites. Radiographic evidence of osteomy- IN) is widely used in human patients for diagnosis of histoplas-
elitis may be present in cats, with associated soft tissue swell- mosis, and urine is the preferred specimen for testing because it
ing and sometimes pathologic fractures. Osteomyelitis is rarely results in a higher sensitivity than when serum is assayed.49 In
described in dogs.30,32 human patients with AIDS and severe PDH, the sensitivity of
the assay is 95% when urine is used, and 86% when serum is
Sonographic Findings used.49 The sensitivity is lower in human patients with pulmo-
Findings on abdominal sonographic examination of dogs with nary histoplasmosis, but use of a combination of urine antigen
histoplasmosis include hepatic enlargement and hypoecho- and serum antigen increases sensitivity to 83%.50 False positives
genicity, abdominal lymphadenopathy, splenomegaly, and asci- occur in people with other disseminated mycoses, including
tes. A thickened intestinal wall may be present with disruption blastomycosis, penicilliosis, and coccidioidomycosis,51,52 so the
of bowel wall architecture. Adrenomegaly, abdominal lymph-
adenopathy, and occasionally renomegaly have been described
in cats.14

Endoscopy
Colonoscopic findings in dogs with colonic histoplasmosis
include irregularity, ulceration, and increased granularity and
friability of the colonic wall (Figure 61-6).

Microbiologic Tests
Diagnostic assays currently available for histoplasmosis in dogs
and cats are described in Table 61-1.

Cytologic Examination
Cytologic examination of aspirates or impression smears of
affected tissues (e.g., fine-needle aspirates of liver, spleen,
lymph nodes, lung, bone marrow), rectal scrapings, or Cytospin
preparations of lung wash specimens or body fluids (e.g., CSF,
synovial fluid, ascites, or pleural effusion) usually reveals pyo-
granulomatous or granulomatous inflammation. H. capsulatum
yeasts can be seen extracellularly and intracellularly (usually
within mononuclear phagocytes). Most extracellular organ-
isms are probably an artifact of smear preparation. Occasion-
ally one or more organisms are seen in circulating monocytes,
neutrophils, or eosinophils in severely ill animals with dissemi- FIGURE 61-6  Colonoscopy image from a dog with disseminated histoplasmosis with
nated disease (Figure 61-7). Cytologic evidence of fungemia was large intestinal involvement. The mucosa is thickened and granular. (Courtesy Dr. Michael
detected in nearly 20% of affected cats in one study.13 The bone Willard, Texas A&M University.)
592 SECTION 3  Fungal and Algal Diseases

TABLE 61-1
Diagnostic Assays Currently Available for Histoplasmosis in Dogs and Cats
Assay Specimen Type Target Performance
Cytologic Aspirates of affected Histoplasma capsulatum Organisms are usually present but, in the absence of
examination tissues, bone marrow as- organisms (usually in experience, may be confused with protozoa such
pirates, sometimes blood large numbers) within as Leishmania or other fungi such as Sporothrix
smears or buffy coat mononuclear phago- schenckii. False negatives can occur in animals with
preparations, body fluids cytes and granulocytes very chronic clinical manifestations.
Fungal culture Aspirates or biopsies of af- H. capsulatum Rarely indicated. Sensitive and specific and may be
fected tissues, body fluids, required for animals with chronic histoplasmosis
whole blood when cytologic examination is negative. Risk of
laboratory-acquired infections. Slow turnaround
time (several weeks of incubation may be required).
Antibody serol- Serum Antibodies to Positive test results indicate exposure but do not
ogy (complement H. capsulatum indicate active infection or correlate with clinical
fixation or gel im- disease. Negative test results occur commonly in
munodiffusion) animals with histoplasmosis (low sensitivity).
Antigen assay Urine, serum H. capsulatum antigen Sensitivity and specificity require further evaluation
(ELISA) in dogs and cats, although the assay appears to be
sensitive and specific in cats when urine is tested.
Cross-reactivity with other fungal pathogens may
occur, so positive results are not diagnostic for his-
toplasmosis. Cross-reactivity is seen with blastomy-
cosis, a systemic fungal infection with an overlap-
ping geographic range.
Real-time PCR See fungal culture H. capsulatum DNA Not yet validated in adequate numbers of dogs or
assays cats with histoplasmosis; usefulness requires further
evaluation.
Histopathology Biopsy specimens from H. capsulatum yeasts Yeasts can be difficult to find in chronic infections.
affected tissues Special stains may assist organism detection.

assay must always be used in conjunction with other diagnostic The sensitivity of one assay was 73% when compared with cul-
assays in order to determine the identity of the infecting fungal ture of clinical specimens.55 PCR assays have not been widely
pathogen. Antigen levels become undetectable with successful applied to the diagnosis of histoplasmosis in dogs and cats; fur-
treatment.53 The antigen assay has also been used for the diag- ther studies are required to determine their usefulness.
nosis of histoplasmosis in cats and dogs.37,54 In one study, the
urine of 17 of 18 cats with confirmed histoplasmosis was posi- Pathologic Findings
tive, whereas none of 26 cats with other diagnoses were posi- Gross pathologic findings in dogs and cats with histoplasmosis
tive.54 Additional studies that evaluate larger numbers of cats consist of miliary nodules or larger focal lesions in the lungs and
and dogs with and without confirmed histoplasmosis, as well other organs, and enlarged lymph nodes, which may be mineral-
as those with other fungal infections, are required to further ized in dogs with chronic disease. In dogs, the intestinal tract may
establish the sensitivity and specificity of this assay. be thickened and focal or diffuse mucosal hemorrhage may be
present. The liver and spleen may be enlarged with rounded bor-
Fungal Culture ders, and the liver may be mottled with a reticulated appearance.
H. capsulatum can be isolated from clinical specimens on rou- Pleural and peritoneal effusions are occasionally described.45
tine fungal media. There is a risk of laboratory-acquired infec- Histopathology yields pyogranulomatous or granulomatous
tion when the organism grows in the mycelial form on artificial inflammation with intralesional yeasts in a variety of organs
media, so culture should be performed only if necessary, and (Figure 61-8). Lymphocytes and plasma cells may be present,
the laboratory should be warned of the possibility of a dimor- and in some animals with chronic histoplasmosis, histologic
phic fungal infection, so that appropriate precautions are taken. evidence of fibrosis or (in dogs) mineralization is apparent.37,45
Although most cultures are positive within 2 or 3 weeks, growth Special stains such as Gomori’s methenamine silver or periodic
may require up to 6 weeks of incubation. The organism is iden- acid–Schiff can aid detection of the yeasts in tissues.
tified based on morphology (see Figure 61-2).
Treatment and Prognosis
Molecular Diagnosis Using the Polymerase Chain Reaction
Real-time PCR assays have been developed for rapid detec- Antimicrobial Treatment
tion of H. capsulatum in clinical specimens from affected Antifungal drugs with activity against H. capsulatum include the
humans55,56 but currently are not used routinely for diagnosis. azole antifungal drugs ketoconazole, itraconazole, and fluconazole
CHAPTER 61  Histoplasmosis 593

FIGURE 61-8  Histopathology of the liver of a 5-month-old intact male redbone


coonhound with disseminated histoplasmosis. There is severe granulomatous inflamma-
tion with massive numbers of intrahistiocytic yeast organisms.

A Deoxycholate or lipid-complexed amphotericin B should


be used initially to treat dogs or cats with severe acute pulmo-
nary, acute disseminated, or CNS disease, after which treatment
should be continued with itraconazole. If funds or patient fac-
tors do not permit itraconazole treatment, fluconazole can be
used as a second-line agent.
The length of treatment and prognosis depend on the
severity and chronicity of disease and the immune status of
the host. At least 6 months of treatment is generally required.
Many animals require treatment for 12 months or longer, and
those with chronic disease may require treatment for more
than 2 years. The decision to discontinue treatment should
be based on lesion resolution (which may require serial imag-
ing studies). Resolution of positive urine antigen titers might
also be used to decide when to discontinue treatment, but fur-
ther study is required to determine whether serial monitoring
of H. capsulatum antigenuria is useful in making treatment
B decisions.
FIGURE 61-7  A, Blood smear from a dog with Histoplasma capsulatum fungemia. Supportive Treatment
Three organisms can be seen in a circulating neutrophil (arrow). Wright’s stain, 1000×
magnification. B, Fine-needle aspirate cytology from the right popliteal lymph node of Other supportive treatments that may be required depend on
the cat in Figure 61-5, A. There is pyogranulomatous inflammation with large numbers the extent of infection and include intravenous crystalloid
of intracellular yeast organisms with morphology consistent with that of H. capsulatum. fluids, supplemental oxygen, blood transfusions for severely
Wright’s stain, 1000× magnification. (Courtesy Angela Royal, Louisiana State University.) anemic animals, nutritional support, antiemetics, and medi-
cations to manage consequences of hepatic failure such as
hepatic encephalopathy. Topical glucocorticoids and atropine
may be required for animals with uveitis, and enucleation may
(see Chapter 9 for more information and dosing for antifungal be required to manage persistent ocular infection and pain.
drugs). Itraconazole and fluconazole are preferred to ketoconazole The role of systemic glucocorticoids for treatment of chronic
since they are more effective and less likely to cause toxicity.17 In manifestations of histoplasmosis is controversial, and they
human patients, itraconazole has consistently been more effective are not recommended for treatment of active or disseminated
than fluconazole. An increased efficacy of itraconazole when com- histoplasmosis.
pared with fluconazole is likely in dogs and cats as well, although
in the authors’ experiences, some animals may be cured with flu- Immunity and Vaccination
conazole monotherapy. Limited data suggest that posaconazole
may be an effective salvage treatment in human patients with Immunity to H. capsulatum is dependent on a Th1 immune
histoplasmosis that fail to respond to treatment with other anti- response. Production of IFN-γ activates macrophages to destroy
fungal drugs, including fluconazole, itraconazole, amphotericin B, the fungus.59 There is currently no vaccine for prevention of
and voriconazole.57 Development of resistance to fluconazole and histoplasmosis, but there is interest in development of a vaccine
voriconazole has been documented during treatment of human for at-risk human patients because of increasing incidence of
patients with fluconazole, but not with posaconazole.58 the disease.
594 SECTION 3  Fungal and Algal Diseases

Prevention PDH.20 Complications of pulmonary histoplasmosis include


granulomatous mediastinitis (characterized by enlargement and
Prevention of histoplasmosis involves avoidance of bat caves sometimes calcification of mediastinal lymph nodes, with com-
and areas previously inhabited by avian species in endemic pression of adjacent structures) or mediastinal fibrosis (char-
areas. Nevertheless, histoplasmosis has occurred in animals acterized by fibrosis of caseous mediastinal lymph nodes). The
housed exclusively indoors, and widespread exposure and clinical picture of PDH in humans is similar to that described in
recovery likely occurs in endemic areas. dogs; rare clinical manifestations include meningitis, endocar-
ditis, and vascular infections.20,49 Humans with AIDS or those
Public Health Aspects treated with immunosuppressive drugs (such as tumor necrosis
factor antagonists) are at risk for severe PDH, and smokers are
As many as 80% of young adults in endemic areas have been pre- predisposed to chronic pulmonary histoplasmosis. Itraconazole
viously infected by H. capsulatum, but only 1% develop symp- prophylaxis is considered for immunosuppressed patients who
toms.49 Risk factors identified for human histoplasmosis include are at risk for development of the disease.20
exposure to soil disrupted as a result of excavation, exploration Transmission of H. capsulatum from dogs or cats to humans
of caves inhabited by bats, or renovation of buildings inhabited has not been reported, but disease in dogs and cats may signal the
by birds or bats. Males are predisposed in a 4:1 ratio.20 Several potential for human infection as a result of exposure to the same
forms of the disease have been described, which include acute source of infection; outbreaks have been described that involved
pulmonary histoplasmosis, chronic pulmonary histoplasmosis both humans and dogs.60 The bodies of deceased pets with his-
(which may be cavitary or noncavitary), and acute or chronic toplasmosis should be disposed of promptly and by cremation.

CANINE CASE EXAMPLE T = 102.2°F (39.0°C), HR = 140 beats/min, RR = 50 breaths/min,


mucous membranes were moist and pink.
Eyes, Ears, Nose, and Throat: Moderate conjunctivitis was
Signalment: “Murphy,” a 5-month-old intact male redbone present. There was a mild bilateral serous nasal discharge.
coonhound from northern California. No other abnormalities were detected.
History: Murphy was evaluated at the University of California, Musculoskeletal: Appeared small in stature, BCS 3/9.
Davis, Veterinary Medical Teaching Hospital for a 1-week Cardiovascular, Respiratory and Lymph Nodes: No clinically
history of increased respiratory effort, hypersalivation, a significant abnormalities were detected.
single episode of vomiting, and lethargy. After the first day Abdominal Palpation: Cranial organomegaly was present.
of illness he was taken to a local veterinary clinic where fever Laboratory Findings:
(103.2°F or 39.6°C), pale mucous membranes, and a cranial Blood Glucose: 70 mg/dL.
abdominal mass were noted on physical examination. Plasma Fasting Ammonia Concentration: 509 µg/dL
Abdominal radiographs revealed moderate hepatomegaly (0-92 µg/dL).
and mild splenomegaly. He was treated with vitamin K, Canine Distemper Virus Direct Immunofluorescent   Anti­
a single dose of prednisone (2 mg/kg SC), and cephalexin body (Urine Sediment): Negative.
(20 mg/kg PO q8h). Considerable improvement in his CBC:
mentation was noted. However, after 5 days, anorexia, HCT 30.2% (40%-55%)
hypersalivation, vomiting, and liquid diarrhea occurred, MCV 61 fL (65-75 fL)
and he returned to the local veterinary clinic, where he was MCHC 36.1 g/dL (33-36 g/dL)
hospitalized and treated with intravenous fluids. The following Reticulocytes 49,200/µL (7000-65,000/µL)
day, obtundation and facial twitching developed, and Murphy WBC 22,000 cells/µL (6000-13,000 cells/µL)
was referred for further diagnostics and treatment. Neutrophils 10,340 cells/µL (3000-10,500 cells/µL)
Other Medical History: Murphy had been coughing a week Band neutrophils 7040 cells/µL
before he became systemically ill, but this subsequently Metamyelocytes 880 cells/µL
resolved. He was treated for a roundworm infection at that Lymphocytes 1100 cells/µL (1000-4000 cells/µL)
time, which was diagnosed by fecal flotation. Murphy was Monocytes 1100 cells/µL (150-1200 cells/µL)
obtained from a breeder in Oklahoma at 4 months of age. He Eosinophils 880 cells/µL (0-1500 cells/µL)
had always appeared “slow” to the owner, his appetite had Platelets 28,000/µL (150,000-400,000 platelets/µL).
never been very good, and he seemed unthrifty. The other Moderately toxic neutrophils and markedly toxic band neu-
puppies in the litter were apparently healthy. There was no trophils and metamyelocytes were present. Occasional
known exposure to toxins. The breeder administered his first organisms with morphology consistent with Histoplasma
vaccines, and he was revaccinated for distemper, parvovirus, capsulatum were seen within both neutrophils and mono-
adenovirus, parainfluenza virus, and Leptospira by the local cytes. The myeloid series was shifted to the myelocyte stage
veterinarian when he was acquired. with rare progranulocytes noted. Rare nucleated erythro-
Physical Examination: cytes (metarubricytes) were also present. Many lympho-
Body Weight: 12.2 kg. cytes were reactive. Numerous macroplatelets were noted.
General: Obtundation was noted; the dog was in lateral Serum Chemistry Profile:
recumbency and apparently unaware of his surroundings. Sodium 143 mmol/L (145-154 mmol/L)
Facial twitching occurred throughout the examination. Potassium 4.7 mmol/L (4.1-5.3 mmol/L)
CHAPTER 61  Histoplasmosis 595

Chloride 108 mmol/L (105-116 mmol/L) Treatment: Murphy was hospitalized in the intensive care
Bicarbonate 18 mmol/L (16-26 mmol/L) unit and treated with intravenous crystalloids (lactated
Phosphorus 7.7 mg/dL (3.0-6.2 mg/dL) Ringer’s solution with 25 mEq/L KCl and 2.5% dextrose,
Calcium 10.3 mg/dL (9.9-11.4 mg/dL) 75 mL/h), plasma, vitamin K, and heparin (100 units/
BUN 18 mg/dL (8-31 mg/dL) kg SC q8h). The hookworm infection was treated with
Creatinine 0.4 mg/dL (0.8-1.6 mg/dL) fenbendazole (50 mg/kg PO q24h for 3 days). Seizures were
Glucose 117 mg/dL (70-118 mg/dL) controlled with diazepam as needed and phenobarbital
Total protein 5.8 g/dL (5.4-7.4 g/dL) (2 mg/kg IM q12h). Hepatic encephalopathy was treated
Albumin 2.1 g/dL (2.9-4.2 g/dL) with a warm water enema followed by a 3 mL/kg lactulose
Globulin 3.7 g/dL (2.3-4.4 g/dL) enema, then oral lactulose (1 mL/kg PO q8h). Profuse
ALT 31 U/L (19-70 U/L) watery diarrhea ensued. Treatment with lipid-complexed
AST 217 U/L (15-43 U/L) amphotericin B was then initiated (2 mg/kg IV over 2 hours,
ALP 195 U/L (15-127 U/L) every other day). Initially, clinical signs improved, and
GGT 24 U/L (0-6 U/L) Murphy appeared brighter and began eating small amounts
Cholesterol 363 mg/dL (135-345 mg/dL) of a reduced protein diet. A CBC showed a hematocrit of 30%,
Total bilirubin 1.1 mg/dL (0-0.4 mg/dL). 19,988 neutrophils/µL, 263 band neutrophils/µL with slight
Urinalysis (Cystocentesis): SGr 1.044; pH 5.0, 1+ bilirubin, toxicity, 2367 lymphocytes/µL, 2367 monocytes/µL, 1315
no hemoprotein, no glucose, no protein, 0-1 RBC/HPF, rare eosinophils/µL, and 65,000 platelets/µL. No Histoplasma
WBC/HPF; no crystals, casts, or bacteria seen. organisms were seen. The results of a biochemistry profile
Coagulation Panel: PT 10.7 s (7.5-10.5 s), PTT 24.4 s (9-12 s), were similar to those reported at hospitalization, but
fibrin degradation products > 40 µg/mL (<10 µg/mL). electrolytes and glucose had normalized and total bilirubin
Imaging Findings: was 2.6 mg/dL.
Thoracic Radiographs: There was a diffuse bronchointerstitial On day 4 of hospitalization, Murphy developed increased respi-
pulmonary pattern, especially in the region of the cranial ratory effort, weakness, and a head tremor. Thoracic radio-
right middle lung lobe. The heart and pulmonary vasculature graphs showed a diffuse miliary interstitial infiltrate in the
were slightly small. In the viewable abdomen, the liver was perihilar region and in the region of the right cranial lung
moderately enlarged. lobe, which was slightly more severe than that seen at the
Abdominal Ultrasound: Severe hepatomegaly was present time of hospitalization. By day 7 of hospitalization, he re-
with normal hepatic echogenicity. A large, folded spleen and mained extremely weak and inappetent, and the owners
sublumbar and mesenteric lymphadenomegaly were present. elected euthanasia because of financial limitations. At nec-
Microbiologic Testing: ropsy, there was severe hepatomegaly and splenomegaly;
Fecal Flotation: Positive for Ancylostoma ova. the lungs were firm and mottled dark red to pink, and cut
Aspiration Cytology (Ultrasound-guided Liver Aspirate): surfaces oozed white froth and blood. Histopathology re-
The specimen was bloody and moderately cellular with vealed severe, diffuse, granulomatous inflammation with
numerous clusters of platelets and inflammatory cells. intrahistiocytic yeasts in the lungs, liver, spleen, bone mar-
Occasional small clusters of hepatocytes were noted, row, sublumbar lymph nodes, and tracheobronchial lymph
which showed mild to moderate vacuolar degeneration. nodes (see Figure 61-8). In the kidneys there was severe,
A few hepatocytes contained intracytoplasmic blue-green multifocal, granulomatous interstitial nephritis with intra-
pigment consistent with bile. Inflammatory cells consisted histiocytic yeasts. Multifocal granulomatous inflammatory
primarily of activated macrophages that contained lesions with intrahistiocytic yeasts were present in the pan-
abundant intracellular yeast organisms consistent with creas, duodenum, jejunum, thyroid glands, and peritoneal
Histoplasma spp. Lower numbers of nondegenerate fat. Within the brain there was minimal, focal, granuloma-
neutrophils, mixed lymphocytes, and occasional eosinophils tous meningitis.
and basophils were also found. Interpretation: Histoplasma Comments: In this dog, the diagnosis of disseminated
infection with marked pyogranulomatous inflammation. histoplasmosis with fungemia was straightforward because
Mild to moderate hepatic vacuolar degeneration and organisms were seen in a peripheral blood smear. The dog
cholestasis. likely acquired infection in Oklahoma where the fungus is
CSF Analysis (Cisternal): The fluid was clear and colorless. endemic. Overwhelming infection suggested a possible
The protein concentration was 17 mg/dL (reference range, underlying immunodeficiency, although severe disease
<25 mg/dL). There was fewer than 1 RBC/µL (reference is common without obvious immunodeficiency in dogs.
range, <1200 RBC/µL) and 25 nucleated cells/µL (reference The dog had evidence of disseminated intravascular
range, <2 cells/µL). The differential cell count consisted coagulation, hepatic encephalopathy, and granulomatous
of 1% neutrophils, 6% lymphocytes, and 93% large meningitis. Had treatment with amphotericin B, azole
mononuclear cells. The cytofuge specimen was cellular, antifungal drugs, and additional supportive care continued,
and large, foamy macrophage-like cells predominated. recovery might have occurred, but the costs of this
Vacuoles within the cytoplasm contained digested debris, treatment without guaranteed success led the owners
but no organisms could be discerned. Low numbers to elect euthanasia. Clearly the burden of intact fungal
of small lymphocytes were also noted. Interpretation: organisms at necropsy was still massive despite several
Mononuclear pleocytosis. treatments with amphotericin B. Even in human patients,
Diagnosis: Acute progressive disseminated histoplasmosis with overall cure rates for H. capsulatum meningitis are no better
suspected Histoplasma meningitis; hookworm infestation. than 50%.
596 SECTION 3  Fungal and Algal Diseases

FELINE CASE EXAMPLE Phosphorus 6.5 mg/dL (3.1-7.5 mg/dL)


Calcium 10.3 mg/dL (8.8-9.3 mg/dL)
BUN 28 mg/dL (16-22 mg/dL)
Signalment: “Shaq,” an 8-month-old intact male domestic Creatinine 1.1 mg/dL (1.7-1.9 mg/dL)
shorthair cat from east central Louisiana. Glucose 125 mg/dL (77-96 mg/dL)
History: Shaq was evaluated at the Louisiana State University Total protein 7.8 g/dL (6.5-7.1 g/dL)
(LSU) Veterinary Medical Teaching Hospital for a 3-week Albumin 3.0 g/dL (2.8-3.1 g/dL)
history of lethargy and intermittent anorexia. Occasional Globulin 4.8 g/dL (37.-4.1 g/dL)
right pelvic limb lameness had been noted. He was taken to ALT 77 U/L (24-44 U/L)
a local veterinary clinic, where fever and an enlarged right AST 16 U/L (0-36 U/L)
popliteal lymph node were noted on physical examination. ALP 41 U/L (33-72 U/L)
A bite wound was suspected at the local veterinarian’s Total bilirubin 0.2 mg/dL (0-0.4 mg/dL).
clinic, and Shaq was treated with an injectable long- Urinalysis: Not performed.
acting cephalosporin and oral amoxicillin. After 2 weeks of Imaging Findings:
continued waxing and waning lethargy, Shaq was brought Thoracic Radiographs: There was a diffuse mild
to the emergency service at LSU. bronchointerstitial pulmonary pattern that was somewhat
Current Medications: Amoxicillin. patchy in the caudodorsal lung fields. A smooth, broad-based
Other Medical History: Shaq was an indoor/outdoor cat soft tissue opaque structure that was sharply marginated by
who has always lived in Louisiana. He ate a commercial the surrounding lung was evident dorsal to sternebrae 3 to
kitten food and was vaccinated at 3 to 4 months of age, 4; this was consistent with sternal lymphadenopathy. In the
which included an FIV vaccination. FeLV and FIV infection visible abdomen, the spleen was moderately enlarged (see
status was unknown. Figure 61-5, A).
Physical Examination: Abdominal Ultrasound: A large spleen was present with
Body Weight: 3.2 kg. diffusely heterogenous echogenicity.
General: The cat was quiet but alert and responsive. His Microbiologic Testing:
mucous membranes were pink and moist, and his CRT was Fecal Flotation: Negative.
less than 2 s. Rectal temperature was 104.9°F (40.5°C), and FeLV/FIV ELISA Serology: Negative for FeLV antigen, positive
heart rate was 216 beats/min. for FIV antibody.
Eyes, Ears, Nose, and Throat: Normal; no fundic examination Aspiration Cytology (Popliteal Lymph Node Aspirate):
abnormalities were noted. Smears contained large numbers of adequately preserved
Musculoskeletal: Normal; no pain was noted on palpation of nucleated cells with a low number of erythrocytes. The
the right pelvic limb. The right popliteal lymph node was nucleated cell population consisted of high numbers of
moderately enlarged, movable, and nonpainful. small lymphocytes (75%), low numbers of neutrophils
Cardiovascular: No murmurs or arrhythmias were auscultated, (20%), and highly vacuolated macrophages (5%). The
and a strong pulse quality was present. macrophages contained large numbers of oval, 2- to 3-µm–
Respiratory: Tachypnea was present but no significant diameter yeast that were surrounded by a thin clear halo
abnormalities were detected on auscultation. and had small, eccentric, crescent-shaped purple nuclei.
Abdominal Palpation: Cranial organomegaly (splenomegaly) Interpretation: Pyogranulomatous inflammation with
was present. intracellular Histoplasma spp. yeasts (see Figure 61-7, B).
Lymph Nodes: Other than the right popliteal lymph node, Histoplasma Quantitative Antigen EIA (Urine): 9.3 ng/mL
lymph nodes were palpably normal. (positive).
Laboratory Findings: Diagnosis: Disseminated histoplasmosis.
CBC: Treatment: Shaq was hospitalized in the intensive care unit
HCT 28.6% (30%-48%) and treated with intravenous crystalloids (Normosol-R with
MCV 33 fL (41-57 fL) 25 mEq/L KCl, 8 mL/h) and his temperature was monitored.
MCHC 30.0 g/dL (30-35 g/dL) The fungal infection was treated with itraconazole oral
Reticulocytes 22,100/µL solution (10 mg/kg PO q24h). His temperature fluctuated
WBC 10,300 cells/µL (6000-10,000 cells/µL) between 103.6°F and 105.3°F (39.8°C and 40.7°C) over the
Neutrophils 6500 cells/µL (2500-12,500 cells/µL) next 48 hours, but he maintained a good appetite and
Lymphocytes 3700 cells/µL (1500-7000 cells/µL) was bright and alert. On day 3, Shaq was discharged from
Monocytes 100 cells/µL (0-800 cells/µL) the hospital with instructions to continue the itraconazole
Eosinophils 0 cells/µL (0-1500 cells/µL) treatment. He was still febrile but was bright and alert and
Platelets 515,000/µL (190,000-368,000 eating well. Recommendations were to recheck monthly
platelets/µL) at the local veterinary clinic for exam and chemistry
WBC and RBC morphology were considered normal. panel. A urinary Histoplasma Quantitative Antigen EIA was
Plasma protein 8.2 g/dL (6-7.5 g/dL). repeated after 2 months of treatment and was 2.0 ng/mL,
Serum Chemistry Profile: which was still positive but indicated a probable treatment
Sodium 164 mmol/L (150-165 mmol/L) response. Recheck at 6 months revealed a normal physical
Potassium 3.7 mmol/L (4.0-5.0 mmol/L) examination, thoracic radiograph, CBC, and chemistry
Chloride 126 mmol/L (121-126 mmol/L) panel with the exception of an increased serum ALT activity
CHAPTER 61  Histoplasmosis 597

at 221 U/L (0-90 U/L). It was recommended that the local of infection in cats. The positive result for FIV antibody was
veterinarian repeat the Histoplasma antigen assay and likely due to the previous vaccination. Treatment is probably
discontinue itraconazole if negative. best continued until the result of the urine antigen assay is
Comments: In this cat, a diagnosis of disseminated negative, but if the titer remains low and clinical signs have
histoplasmosis was straightforward because organisms were resolved, discontinuation of itraconazole treatment may be
seen in a lymph node aspirate. If they had not been present appropriate with retesting for antigenuria. An increase in
on cytologic examination of the lymph node aspirate, urine antigen concentration may indicate relapse and need
cytologic examination of a splenic or a bone marrow for further therapy. The increased ALT (221 U/L) was likely due
aspirate would have been most likely to reveal organisms. to itraconazole therapy and did not necessitate alteration in
The vague presentation with mild respiratory signs is typical treatment as long as the cat otherwise appeared normal.

SUGGESTED READINGS 15. Mitchell M, Stark DR. Disseminated canine histoplasmosis: a clini-
cal survey of 24 cases in Texas. Can Vet J. 1980;21:95-100.
Aulakh HK, Aulakh KS, Troy GC. Feline histoplasmosis: a retro- 16. Selby LA, Becker SV, Hayes Jr HW. Epidemiologic risk fac-
spective study of 22 cases (1986-2009). J Am Anim Hosp Assoc. tors associated with canine systemic mycoses. Am J Epidemiol.
2012;48:182-187. 1981;113:133-139.
Cook AK, Cunningham LY, Cowell AK, et al. Clinical evaluation of urine 17. Hodges RD, Legendre AM, Adams LG, et  al. Itraconazole
Histoplasma capsulatum antigen measurement in cats with suspected for the treatment of histoplasmosis in cats. J Vet Intern Med.
disseminated histoplasmosis. J Feline Med Surg. 2012;14:512-515. 1994;8:409-413.
Kroetz DN, Deepe GS. The role of cytokines and chemokines in Histo- 18. Clinkenbeard KD, Cowell RL, Tyler RD. Disseminated histo-
plasma capsulatum infection. Cytokine. 2012;58:112-117. plasmosis in dogs: 12 cases (1981-1986). J Am Vet Med Assoc.
Lin Blache J, Ryan K, Arceneaux K. Histoplasmosis. Compend Contin 1988;193:1443-1447.
Educ Vet. 2011;33:E1-E11. 19. Wolf AM, Green RW. The radiographic appearance of pulmonary
histoplasmosis in the cat. Vet Radiol. 1987;28:34-37.
REFERENCES 20. Deepe GS. Histoplasma capsulatum. In: Mandell GL, Bennett JE,
1. Darling ST. A protozoal general infection producing pseudotuber- Dolin R, eds. Principles and Practice of Infectious Diseases. 7th ed.
cles in the lungs and focal necrosis in the liver, spleen, and lymph Philadelphia, PA: Elsevier; 2010:3305-3318.
nodes. JAMA. 1906;46:1283-1285. 21. Newman SL, Gootee L, Hilty J, et  al. Human macrophages do
2. Johnson LR, Fry MM, Anez KL, et  al. Histoplasmosis infec- not require phagosome acidification to mediate fungistatic/fun-
tion in two cats from California. J Am Anim Hosp Assoc. gicidal activity against Histoplasma capsulatum. J Immunol.
2004;40:165-169. 2006;176:1806-1813.
3. Brilhante RS, Coelho CG, Sidrim JJ, et al. Feline histoplasmosis in 22. Hilty J, George Smulian A, Newman SL. Histoplasma capsulatum
Brazil: clinical and laboratory aspects and a comparative approach utilizes siderophores for intracellular iron acquisition in macro-
of published reports. Mycopathologia. 2012;173:193-197. phages. Med Mycol. 2011;49:633-642.
4. Mavropoulou A, Grandi G, Calvi L, et al. Disseminated histoplas- 23. Emmons CW, Rowley DA, Olson BJ, et al. Histoplasmosis; proved
mosis in a cat in Europe. J Small Anim Pract. 2010;51:176-180. occurrence of inapparent infection in dogs, cats and other animals.
5. Mackie JT, Kaufman L, Ellis D. Confirmed histoplasmosis in an Am J Hyg. 1955;61:40-44.
Australian dog. Aust Vet J. 1997;75:362-363. 24. Goad MEP, Roenick WJ. Osseous histoplasmosis in a cat. Fel
6. Murata Y, Sano A, Ueda Y, et al. Molecular epidemiology of canine Pract. 1983;13:32-36.
histoplasmosis in Japan. Med Mycol. 2007;45:233-247. 25. Tamulevicus AM, Harkin K, Janardhan K, et  al. Disseminated
7. Sanford SE, Straube U. Ontario. Disseminated histoplasmosis in a histoplasmosis accompanied by cutaneous fragility in a cat. J Am
young dog. Can Vet J. 1991;32:692. Anim Hosp Assoc. 2011;47:e36-e41.
8. Tyre E, Eisenbart D, Foley P, et al. Histoplasmosis in a dog from 26. Lamm CG, Rizzi TE, Campbell GA, et  al. Pathology in prac-
New Brunswick. Can Vet J. 2007;48:734-736. tice. Histoplasma capsulatum infections. J Am Vet Med Assoc.
9. Kasuga T, White TJ, Koenig G, et al. Phylogeography of the fungal 2009;235:155-157.
pathogen Histoplasma capsulatum. Mol Ecol. 2003;12:3383-3401. 27. Vinayak A, Kerwin SC, Pool RR. Treatment of thoracolumbar
10. Arunmozhi Balajee S, Hurst SF, Chang L, et al. Multilocus sequence spinal cord compression associated with Histoplasma capsulatum
typing of Histoplasma capsulatum in formalin-fixed paraffin- infection in a cat. J Am Vet Med Assoc. 2007;230:1018-1023.
embedded tissues from cats living in non-endemic regions reveals a 28. Taylor AR, Barr JW, Hokamp JA, et al. Cytologic diagnosis of dis-
new phylogenetic clade. Med Mycol. 2012:Oct 17 [Epub]. seminated histoplasmosis in the wall of the urinary bladder of a cat.
11. Zeidberg LD, Ajello L, Webster RH. Physical and chemical fac- J Am Anim Hosp Assoc. 2012;48:203-208.
tors in relation to Histoplasma capsulatum in soil. Science. 29. Kobayashi R, Tanaka F, Asai A, et al. First case report of histoplas-
1955;122:33-34. mosis in a cat in Japan. J Vet Med Sci. 2009;71:1669-1672.
12. Taylor ML, Hernandez-Garcia L, Estrada-Barcenas D, et  al. 30. Burk RL, Jones BD. Disseminated histoplasmosis with osseous
Genetic diversity of Histoplasma capsulatum isolated from infected involvement in a dog. J Am Vet Med Assoc. 1978;172:1416-1417.
bats randomly captured in Mexico, Brazil, and Argentina, using the 31. Gilor C, Ridgway MD, Singh K. DIC and granulomatous vasculitis
polymorphism of (GA)(n) microsatellite and its flanking regions. in a dog with disseminated histoplasmosis. J Am Anim Hosp Assoc.
Fungal Biol. 2012;116:308-317. 2011;47:e26-e30.
13. Davies C, Troy GC. Deep mycotic infections in cats. J Am Anim 32. Lau RE, Kim SN, Pirozok RP. Histoplasma capsulatum infection in
Hosp Assoc. 1996;32:380-391. a metatarsal of a dog. J Am Vet Med Assoc. 1978;172:1414-1416.
14. Aulakh HK, Aulakh KS, Troy GC. Feline histoplasmosis: a retro- 33. Nishifuji K, Ueda Y, Sano A, et  al. Interdigital involvement in a
spective study of 22 cases (1986-2009). J Am Anim Hosp Assoc. case of primary cutaneous canine histoplasmosis in Japan. J Vet
2012;48:182-187. Med A Physiol Pathol Clin Med. 2005;52:478-480.
598 SECTION 3  Fungal and Algal Diseases

34. Kagawa Y, Aoki S, Iwatomi T, et al. Histoplasmosis in the skin and 48. Miller BM, Waterhouse G, Alford RH, et al. Histoplasma infection
gingiva in a dog. J Vet Med Sci. 1998;60:863-865. of abdominal aortic aneurysms. Ann Surg. 1983;197:57-62.
35. Ueda Y, Sano A, Tamura M, et al. Diagnosis of histoplasmosis by 49. Kauffman CA. Histoplasmosis: a clinical and laboratory update.
detection of the internal transcribed spacer region of fungal rRNA Clin Microbiol Rev. 2007;20:115-132.
gene from a paraffin-embedded skin sample from a dog in Japan. 50. Swartzentruber S, Rhodes L, Kurkjian K, et al. Diagnosis of acute
Vet Microbiol. 2003;94:219-224. pulmonary histoplasmosis by antigen detection. Clin Infect Dis.
36. Sano A, Miyaji M. Canine histoplasmosis in Japan. Nihon Ishinkin 2009;49:1878-1882.
Gakkai Zasshi. 2003;44:239-243. 51. Wheat J, Wheat H, Connolly P, et  al. Cross-reactivity in Histo-
37. Clemans JM, Deitz KL, Riedesel EA, et  al. Retroperitoneal pyo- plasma capsulatum variety capsulatum antigen assays of urine
granulomatous and fibrosing inflammation secondary to fungal samples from patients with endemic mycoses. Clin Infect Dis.
infections in two dogs. J Am Vet Med Assoc. 2011;238:213-219. 1997;24:1169-1171.
38. Olson GA, Wowk BJ. Oral lesions of histoplasmosis in a dog. Vet 52. Kuberski T, Myers R, Wheat LJ, et al. Diagnosis of coccidioidomy-
Med Small Anim Clin. 1981;76:1449-1451. cosis by antigen detection using cross-reaction with a Histoplasma
39. Huss BT, Collier LL, Collins BK, et al. Polyarthropathy and chorio- antigen. Clin Infect Dis. 2007;44:e50-e54.
retinitis with retinal detachment in a dog with systemic histoplas- 53. Hage CA, Kirsch EJ, Stump TE, et al. Histoplasma antigen clear-
mosis. J Am Anim Hosp Assoc. 1994;30:217-224. ance during treatment of histoplasmosis in patients with AIDS
40. Gwin RM, Makley Jr TA, Wyman M, et  al. Multifocal ocu- determined by a quantitative antigen enzyme immunoassay. Clin
lar histoplasmosis in a dog and cat. J Am Vet Med Assoc. Vaccine Immunol. 2011;18:661-666.
1980;176:638-642. 54. Cook AK, Cunningham LY, Cowell AK, et al. Clinical evaluation
41. Meadows RL, MacWilliams PS, Dzata G, et al. Diagnosis of his- of urine Histoplasma capsulatum antigen measurement in cats
toplasmosis in a dog by cytologic examination of CSF. Vet Clin with suspected disseminated histoplasmosis. J Feline Med Surg.
Pathol. 1992;21:122-125. 2012;14:512-515.
42. Schaer M, Johnson KE, Nicholson AC. Central nervous system dis- 55. Martagon-Villamil J, Shrestha N, Sholtis M, et al. Identification of
ease due to histoplasmosis in a dog: a case report. J Am Anim Hosp Histoplasma capsulatum from culture extracts by real-time PCR.
Assoc. 1983;19:311-316. J Clin Microbiol. 2003;41:1295-1298.
43. Clinkenbeard KD, Cowell RL, Tyler RD. Disseminated histo- 56. Babady NE, Buckwalter SP, Hall L, et  al. Detection of Blasto-
plasmosis in cats: 12 cases (1981-1986). J Am Vet Med Assoc. myces dermatitidis and Histoplasma capsulatum from culture isolates
1987;190:1445-1448. and clinical specimens by use of real-time PCR. J Clin Microbiol.
44. Carakostas MC, Miller RI, Gossett KA. Clinical laboratory 2011;49:3204-3208.
evaluation of deep mycotic diseases in dogs. J Small Anim Pract. 57. Restrepo A, Tobon A, Clark B, et al. Salvage treatment of histo-
1984;25:687-693. plasmosis with posaconazole. J Infect. 2007;54:319-327.
45. Kowalewich N, Hawkins EC, Skowronek AJ, et al. Identification 58. Wheat LJ, Connolly P, Smedema M, et al. Activity of newer triazoles
of Histoplasma capsulatum organisms in the pleural and peritoneal against Histoplasma capsulatum from patients with AIDS who
effusions of a dog. J Am Vet Med Assoc. 1993;202:423-426. failed fluconazole. J Antimicrob Chemother. 2006;57:1235-1239.
46. Clinkenbeard KD, Cowell RL, Tyler RD. Identification of Histo- 59. Kroetz DN, Deepe GS. The role of cytokines and chemokines in
plasma organisms in circulating eosinophils of a dog. J Am Vet Histoplasma capsulatum infection. Cytokine. 2012;58:112-117.
Med Assoc. 1988;192:217-218. 60. Davies SF, Colbert RL. Concurrent human and canine his-
47. Burk RL, Corley EA, Corwin LA. The radiographic appearance of toplasmosis from cutting decayed wood. Ann Intern Med.
pulmonary histoplasmosis in the dog and cat: a review of 37 case 1990;113:252-253.
histories. J Am Vet Radiol Soc. 1978;19:2-7.
CHAPTER 62

Cryptococcosis
Jane E. Sykes and Richard Malik

disease transmission from one animal to another has not been


Overview of Cryptococcosis reported. Occasional outbreaks of cryptococcosis in people
First Described: 1894, in a human patient in Germany (inde- and animals may result from exposure to common environ-
pendently by Busse and Buschke)1 mental sources.2,3 Various domestic and native mammals may
be infected, which include cats, dogs, ferrets, horses, camelids,
Causes: Cryptococcus gattii, Cryptococcus neoformans, rarely
goats, sheep, cattle, dolphins, birds, koalas, and other marsu-
other cryptococcal species (a basidiomycete, teleomorph
pials.4-10 The incidence of cryptococcosis in cats is eightfold
Filobasidiella spp.)
higher than that in dogs.11
Affected Hosts: Cats, dogs, humans, and a variety of other The genus Cryptococcus contains at least 19 encapsulated
domestic and wild animal species yeast species. Cryptococcus neoformans and Cryptococcus gat-
Geographic Distribution: Worldwide tii (formerly C. neoformans var. gattii) cause the vast majority
of infections in dogs, cats, and humans. C. gattii has emerged as
Major Mode of Transmission: Inhalation of basidiospores
a pathogen of immunocompetent humans in temperate regions
from the environment
of North America, in particular on the west coast of the United
Major Clinical Signs: Nodular or ulcerative cutaneous lesions, States and in British Columbia, Canada. Other species, particu-
upper respiratory tract signs, chorioretinitis, neurologic larly Cryptococcus laurentii and Cryptococcus albidus, rarely
signs secondary to meningoencephalitis. In dogs, Cryp- cause disease in people and animals, but may be emerging as
tococcus can also disseminate to abdominal organs and a result of immunocompromise.12-14 Cryptococcus magnus
cause gastrointestinal signs such as vomiting, diarrhea, was isolated from the external ear canal of a cat with otitis
and inappetence. externa,15 and C. laurentii has been isolated from the external
Differential Diagnoses: For cats, differential diagnoses for ear canal of dogs.16
cutaneous lesions include other deep mycoses (especially Cryptococcus spp. are dimorphic, basidiomycetous fungi.
sporotrichosis), mycobacteriosis, nocardiosis, neoplasia The life cycle of the fungus is thought to involve growth in host
(especially squamous cell carcinoma), herpetic derma- tissues as an encapsulated haploid budding yeast, combined
titis and eosinophilic granuloma complex. Differential with the ability to undergo a transition to a filamentous form
diagnoses for suspected cryptococcal rhinitis include (or teleomorph) in the environment. Two mating types of Cryp-
nasal lymphoma, foreign bodies, sinonasal/sino-orbital tococcus spp. exist, α and a. The α-type is vastly more preva-
aspergillosis, oronasal fistulae and neutrophilic rhinitis. lent in environmental and clinical samples. Under appropriate
For ocular and central nervous system lesions, differential laboratory conditions, the two mating types fuse and adopt a
diagnoses include neoplasia (especially lymphoma), toxo- dikaryotic filamentous state, also known as the perfect state.
plasmosis, feline infectious peritonitis, and otogenic bac- This is followed by production of basidia, which are small, club-
terial meningitis. For dogs, differential diagnoses include shaped structures on which basidiospores form (Figure 62-1).17
other disseminated mycoses (especially histoplasmo- Cells of the α mating type can also undergo asexual reproduc-
sis and blastomycosis), mycobacteriosis, actinomycosis tion via a process known as monokaryotic fruiting, which also
and nocardiosis, other causes of rhinitis (such as foreign involves filamentation and spore formation.17-19 Although they
bodies, nasal aspergillosis, oronasal fistulas, lymphoplas- have never been observed in nature, basidiospores (or possibly
macytic rhinitis, rhinosporidiosis), neoplasia (especially desiccated yeasts) are thought to be the main form that, when
round cell neoplasia), toxoplasmosis, neosporosis, severe inhaled, gives rise to mammalian disease.20 In animal tissues,
inflammatory bowel disease, and pancreatitis. basidiospores convert to the yeast form, which is round to oval
with a variably sized capsule. The capsule is primarily (90% to
Human Health Significance: Human infections occur as a 95%) composed of a polysaccharide known as glucuronoxy-
result of exposure to organisms in the environment and are lomannan (GXM), with smaller amounts of galactoxyloman-
often associated with underlying immune compromise. nan. The capsule enlarges after infection of mammalian hosts
and protects the fungus from phagocytosis and environmental
insults such as desiccation. Within tissues, Cryptococcus repro-
Etiology and Epidemiology duces by forming one or two daughter cells (buds) that are con-
nected to the parent cell by a narrow base (Figure 62-2).
Cryptococcosis is an important fungal infection of people and Historically, five serotypes of Cryptococcus (A, B, C, D, and
animals worldwide and the most common systemic mycosis AD) were recognized on the basis of antigenic differences in
of cats. The infection is acquired from the environment, and the capsular polysaccharide. Based on the results of molecular

599
600 SECTION 3  Fungal and Algal Diseases

A B
FIGURE 62-1  Life cycle of Cryptococcus spp. A, Spores result from either mating between cells of opposite mating types (MATa or MATα) and subsequent formation of dikaryotic
hyphae or unisexual mating of MATα cells (monokaryotic fruiting). In both cases, sexual development within the filamentous form results in meiosis and basidiospore formation. The spores
germinate to produce haploid, yeast-like cells that divide by budding. B, Spores, and potentially desiccated yeast cells, are inhaled. Germination of spores or vegetative growth of yeast cells
in the upper or lower respiratory tract results in the proliferation of budding yeasts. Dissemination to a variety of tissues can then occur, but especially to the lymph nodes, nasal cavity, skin,
and CNS. (Modified from Kronstad JW, Attarian R, Cadieux B, et al. Expanding fungal pathogenesis: Cryptococcus breaks out of the opportunistic box. Nat Rev Microbiol 2011;9:193-203.
Basidiospore image courtesy Patrik Inderbitzen, University of California, Davis.)

cats have been predisposed in Australian studies but were not


found to be at risk in the United States.11,23 Similarly, German
shepherd dogs, Doberman pinschers, and great Danes appear to
be predisposed in Australia, whereas in the United States, Amer-
ican cocker spaniel dogs are strongly predisposed.5,11,22,24,25
Most cats and dogs with cryptococcosis do not have iden-
tifiable underlying immunosuppressive illness. The prevalence
of retrovirus infection in cat populations with cryptococcosis
does not differ from that in cat populations without crypto-
coccosis.11,22 However, some cats with cryptococcosis that are
co-infected with FeLV appear to respond more slowly to treat-
ment and may be more likely to have relapses.26 Cryptococcosis
is occasionally reported in cats treated with immunosuppres-
sive therapy or chemotherapy for malignancy.11,27 Concurrent
opportunistic infections have been reported in other cats (e.g.,
toxoplasmosis), as well as in dogs (e.g., neosporosis or papillo-
FIGURE 62-2  CSF cytology from an 8-year-old male neutered domestic shorthair cat matosis), suggesting underlying immunodeficiency.11,28
with cryptococcal meningitis. Large numbers of budding cryptococcal yeasts are present.
Cryptococcus neoformans
typing methods such as PCR fingerprinting and multilocus C. neoformans has a worldwide distribution. It primarily infects
sequence typing, cryptococcal strains from around the world human patients who are immunocompromised, especially AIDS
are divided into eight major molecular types: VNI and VNII patients. The realized environmental niche for C. neoformans
(C. neoformans serotype A); VNIII (C. neoformans serotype is weathered bird (especially pigeon) guano, in which it mates
AD); VNIV (C. neoformans serotype D), and VGI, VGII, VGIII, prolifically.29 It can also be found in decaying plant matter in
and VGIV (C. gattii, serotypes B and C).21 Hybrid strains also hollows of certain trees.30 The organism can remain viable for
exist. Differences in epidemiology, pathogenicity, clinical fea- at least 2 years in environments such as pigeon lofts. The major-
tures, and drug susceptibility have been associated with each ity of human cryptococcal infections result from infection with
species and molecular type. C. neoformans var. grubii (VNI). C. neoformans var. grubii is
Cats of all ages may be affected,22 but young adult cats the most common Cryptococcus species isolated from dogs and
appear to be at increased risk. In dogs, cryptococcosis is diag- cats in southeastern Australia and accounts for more than 70%
nosed predominantly in young adult dogs. In one study, the of infections.22,23,25 Across the United States, most dogs with
median age of cats and dogs was 6 and 4 years, respectively.11 cryptococcosis are infected with C. neoformans, but infection
No clear sex predisposition exists. Siamese, birman, and ragdoll of cats with this species is rare.11,31
CHAPTER 62  Cryptococcosis 601

Cryptococcus gattii
C. gattii is endemic in Australia, New Zealand, Papua New
Guinea, Southeast Asia, parts of Latin America, California, Mex-
ico, Hawaii, Central and South Africa, and, to a lesser extent,
certain parts of Europe (Austria, Germany, France, Italy, Greece,
and Spain).32,33 Since 1999, an epidemic of infections with
C. gattii has occurred in humans and domestic and wild ani-
mals in British Columbia in Canada, especially Vancouver
Island, and the Pacific Northwest of the United States.34-36
The main molecular types identified in this epidemic have been
molecular types VGIIa and, to a much lesser extent, VGIIb
and, in Oregon, also VGIIc. C. gattii molecular type VGII
can be found on living tree bark (Garry oak, maple, cedar,
and pine) and in air, and has also been found in freshwater
and seawater. Proximity to areas with soil disturbance was a
risk factor for dogs and cats.37 In California, almost all cats
are infected with C. gattii VGIII, with occasional VGII infec- FIGURE 62-3  Ultrasound image that shows an enlarged and hypoechoic pancreas
in a 7-year-old female spayed basset mixed-breed dog with systemic cryptococcosis
tions.11,31 The ecologic niche of this molecular type in the
caused by Cryptococcus neoformans var. grubii (molecular type VNI). The dog was evalu-
United States remains unclear. Approximately 25% of affected ated for gastrointestinal signs that were associated with severe cryptococcal pancreatitis
cats are housed exclusively indoors.11 and enteritis. The dog’s lungs, heart, kidneys, liver, mesenteric lymph nodes, peritoneum,
In eastern Australia, 20% to 30% of human, feline, and brain, and meninges were also involved at necropsy.
canine cryptococcosis is caused by C. gattii,23,38 whereas in
western Australia, approximately 50% of cats and dogs are
infected with C. gattii.5 Most C. gattii isolates from eastern Aus- phospholipase, urease, and superoxide dismutase.17,42 Capsular
tralia belong to VGI; VGII predominates in southwestern and polysaccharide is continuously shed into the host’s extracellular
northern Australia.5,39 C. gattii VGI in Australia is exclusively fluid, which includes blood, urine, and CSF. The capsule inhib-
associated with dead plant material in eucalyptus tree hollows. its phagocytosis, depletes complement, and is well known for
Cats in rural environments are at risk of infection. its ability to inhibit effective T cell responses. The ability of C.
In contrast to C. neoformans, C. gattii isolates from the neoformans and C. gattii to grow at mammalian body tempera-
Pacific northwest and British Columbia can infect apparently ture is also an important virulence factor and is lacking in other
immunocompetent humans.40 C. gattii molecular type VGIII species of Cryptococcus.
infections have been reported in immunosuppressed humans in The incubation period can range from 2 to 13 months.45,46
southern California.41 When disease occurs in immunocompe- Exposure and self-limiting infection may occur in some individu-
tent hosts, it is often characterized by the presence of pulmonary als, with reactivation of viable cryptococci in residual granulo-
mass lesions (cryptococcomas) and sometimes intracranial cryp- matous foci months or years later with immunosuppressive drug
tococcomas.42 As a result, pulmonary cryptococcomas are more therapy or illness.42 Because of this delay, a travel history to areas
commonly described in human patients infected with C. gattii where Cryptococcus is highly endemic (such as British Columbia
compared with those infected with C. neoformans.43 or the west coast of the United States) may be present in animals
and humans residing in non-endemic regions. After infection is
Clinical Features established within the lung or nasal cavity, the organism can
spread hematogenously to other sites, particularly the lymph
Signs and Their Pathogenesis nodes, eye, skin, and central nervous system (CNS) (see Figure
Based on the small size of basidiospores, the lung is thought to 62-1). Cryptococcus spp. has a strong tendency to invade the
be the primary site of infection in humans. In cats, dogs, and meninges and typically causes meningitis or meningoencephali-
koalas, the nasal cavity may be the primary site of infection (see tis. In humans, cerebral cryptococcosis is often associated with
Figure 62-1); significant pulmonary parenchymal involvement increased intracranial pressure, and this probably also occurs
in these species is rare. Colonization of the nasal cavity may be in cats and dogs. Spread of infection to the eye, either along
followed by development of cryptococcal rhinitis in some (but the optic nerves from the brain or as a result of hematogenous
not all) dogs and cats. However, some nasal cavity involvement spread, may result in concurrent cryptococcal optic neuritis and
may result from hematogenous dissemination, because injec- chorioretinitis. In animals with nasal cavity involvement, exten-
tion of Cryptococcus spp. into the carotid artery of cats can sion of infection into the brain may also follow osteomyelitis of
lead to development of lesions on the bridge of the nose. Some the cribriform plate or of the ventral wall of the frontal sinus.
localized cutaneous lesions may result from direct cutaneous Middle ear involvement can also occur.47 Infection also often
inoculation (e.g., after a cat scratch), but skin lesions are usu- spreads from the nasal cavity to the mandibular lymph nodes.
ally interpreted as the result of dissemination or extension from Multifocal cutaneous involvement in cats and dogs reflects
the nasal cavity. hematogenous dissemination from the primary site of infection,
The pathogenesis of cryptococcosis depends on the size of as do lesions in bone (e.g., digits) or periarticular soft tissues.
the inoculum, virulence of the cryptococcal strain, and host Dogs infected with C. neoformans often develop widely dis-
immunity. Cryptococcus has been aptly described as “a sugar- seminated disease, with involvement of sites such as the gastro-
coated killer with designer genes.”44 Cryptococcus has several intestinal system, pancreas, mesenteric lymph nodes, kidneys,
important virulence factors, including its polysaccharide cap- myocardium, and thyroid gland (Figure 62-3).11,24,48,49 In con-
sule, laccase (which makes melanin), and other enzymes such as trast, dogs infected with C. gattii molecular type VGII in the
602 SECTION 3  Fungal and Algal Diseases

United States tend to have isolated caudal nasal cavity crypto- Lower respiratory tract signs such as tachypnea are uncom-
coccomas, which can extend through the cribriform plate into mon but can occur as a result of pleural effusion or severe medi-
the brain (Figure 62-4). astinal lymphadenomegaly. Neurologic signs vary depending
on the location of lesions in and around the CNS (see Physical
Clinical Features in Cats Examination Findings), and, rarely, cats with CNS involvement
Cryptococcosis is often a chronic infection in cats, and affected lack obvious neurologic signs. Optic neuritis and chorioretinitis
cats are usually otherwise well but occasionally have mild develop in as many as one third of affected cats and almost
lethargy and inappetence. Fever is rare, and when it occurs, always indicate CNS involvement.
it tends to be low-grade. Upper respiratory tract signs due to Other signs include peripheral lymphadenomegaly (which
nasal cavity involvement are common, sometimes with associ- may be unassociated with skin lesions and is often asymmet-
ated mandibular lymphadenopathy. Single or multifocal ulcer- ric), lameness due to osteomyelitis or cryptococcal arthritis, and
ated or nonulcerated cutaneous masses may also be present swollen digits. Rarely, involvement of the abdominal lymph
(Figure 62-5). nodes, kidneys, spleen, liver, thyroid, and salivary glands occur.

B C
FIGURE 62-4  Caudal nasal cryptococcosis in dogs infected with Cryptococcus gattii. A, CT scan showing a large caudal nasal cavity cryptococcoma at necropsy that invaded the cribriform
plate and retrobulbar space in a 5-year-old Doberman pinscher infected with C. gattii molecular type VGIIa. There is a palisading periosteal response along the lateral margin of the right frontal and
lacrimal bones. B and C, Postcontrast magnetic resonance images of the head of a 2-year-old intact male malamute with a 1-week history of abnormal behavior and seizures. Cryptococcus gattii
was isolated from the nasal cavity. A large, contrast-enhancing mass is present in the caudal nasal cavity that invades the cribriform plate and extends into the brain. (Courtesy Dr. Stacey Hoffman.)
CHAPTER 62  Cryptococcosis 603

Clinical Features in Dogs were present in two thirds of affected dogs from ­California.11
In contrast to cats, dogs frequently develop severe dissemi- Dogs can also primarily show signs of gastrointestinal or
nated disease. In one study, 80% of dogs had involvement of pancreatic involvement, such as vomiting, diarrhea, and
multiple anatomic sites, and 50% of dogs had involvement abdominal pain.
of sites other than the nasal cavity, skin, lungs, lymph nodes, Cutaneous involvement is uncommon in dogs but, as in cats,
kidney, eye, and CNS.11 Weight loss, lethargy, and inappe- may be a marker for disseminated disease. Although rarely
tence are common nonspecific findings. Rhinosinusitis from detected antemortem, more than half of dogs have lower respi-
cryptococcosis may be subclinical in dogs, so its incidence ratory involvement at necropsy.11 Other clinical signs that are
may be underestimated.11,25 However, sneezing and muco- infrequently present in dogs include peripheral lymphadeno-
purulent nasal discharge can also occur. Neurologic signs megaly or lameness (due to osteomyelitis or arthritis).

Physical Examination Findings


Cats
Upper respiratory tract signs are common physical examina-
tion findings in cats with cryptococcosis. These include sneez-
ing; snuffling; and mucopurulent, serous, or hemorrhagic
nasal discharge that is unilateral or bilateral. In some cats, a
fleshy mass protrudes from the nostril. Other cats have one or
more firm to fluctuant, ulcerated or nonulcerated, subcutane-
ous swellings over the bridge of the nose. These swellings may
communicate with the frontal sinus or nasal cavity, and palpa-
tion of the lesions may reveal crepitus due to subcutaneous
emphysema. Cats with nasopharyngeal cryptococcosis develop
stertor, stridor, and, occasionally, otitis media. Mandibular
lymphadenopathy is often present. Ulcerated or proliferative
lesions of the buccal mucosa can also be found. Multifocal skin
lesions consist of nodules or masses that are fluctuant to firm,
up to several centimeters in diameter, and may ulcerate (see
Figure 62-5).
Neurologic examination abnormalities in cats with CNS
involvement include obtundation; atypical behavior; slow
menace, palpebral, or pupillary light responses; mydriasis
or anisocoria (Figure 62-6, A); twitching or tremors; and sei-
zures, circling, head pressing, ataxia, paresis, abnormal placing
reactions, head tilt, nystagmus, and spinal pain.9 Fundoscopic
examination may reveal granulomatous chorioretinitis, exuda-
FIGURE 62-5  Multiple ulcerated skin lesions in a cat from northern California with tive retinal detachment, papilledema, evidence of optic neuri-
disseminated cryptococcosis caused by Cryptococcus gattii molecular type VGIII. tis, and/or retinal hemorrhage (Figure 62-6, B). Severe ocular

A B
FIGURE 62-6  Ocular abnormalities in cats with disseminated cryptococcosis. A, Bilateral mydriasis in an 8-year-old male neutered domestic shorthair with cryptococcal menin-
goencephalitis. Fundoscopic examination was within normal limits. B, Granulomatous chorioretinitis and retinal detachment in another cat with cryptococcosis. (Courtesy University of
California, Davis, Veterinary Ophthalmology Service.)
604 SECTION 3  Fungal and Algal Diseases

TABLE 62-1
Diagnostic Assays Currently Available for Cryptococcosis in Dogs and Cats
Assay Specimen Type Target Performance
Cytologic Aspirates of affected tissues, Cryptococcus High sensitivity, but occasionally organisms are not
­examination body fluids, impression ­organisms ­visualized (such as in the CSF of some animals with
smears of skin lesions or CNS cryptococcosis).
biopsies
Fungal culture Aspirates or biopsies of af- Cryptococcus spp. Generally sensitive and specific, but false negatives are
fected tissues, body fluids possible if specimen size is low. Requires several days’
such as CSF and urine incubation. Allows species identification and antifungal
drug susceptibility testing.
Antigen assay Serum, CSF Cryptococcus Sensitivity and specificity exceed 90%. False negatives
(latex agglu- ­polysaccharide can occur in dogs and with localized infections in cats.
tination) antigen False positives are generally of low magnitude (<1:200
and usually <1:16). Titers vary between kits.
Real-time PCR Biopsies, body fluids, aspi- Cryptococcus DNA Not yet validated in adequate numbers of dogs or cats
assays rates with cryptococcosis; usefulness requires further evalu-
ation.
Histopathology Biopsy specimens from af- Cryptococcus spp. High sensitivity, with rare false negatives. Special stains
fected tissues yeasts such as Mayer’s mucicarmine and immunohistochemi-
cal stains facilitate organism detection and identifica-
tion.

lesions or optic neuritis may result in blindness with dilated and moderate nonregenerative anemia; leukocytosis, often with mono-
unresponsive pupils. cytosis; and eosinophilia are the most common findings. Dogs with
disseminated disease may have a neutrophilia with a left shift. The
Dogs serum biochemical profile reflects specific organ involvement in
Many dogs with cryptococcosis are lethargic or obtunded animals with disseminated disease. Occasionally, cryptococcal
because of disseminated disease and/or CNS involvement. When yeasts are found on examination of the urine sediment.
fever occurs, it is typically low grade. Other CNS signs in dogs
include absent or slow menace, gag, pupillary light responses, Cerebrospinal Fluid Examination
nystagmus, mydriasis, head tilt, strabismus, anisocoria, facial The CSF in cats and dogs with neurologic signs due to crypto-
paralysis, paraparesis, abnormal placing reactions, ataxia, cir- coccosis may have an increased protein concentration (usually
cling, seizures, twitching or tremors, reluctance to open the between 10 and 140 mg/dL in cats, but may be several hundred
mouth, absent or decreased segmental reflexes, and, commonly, or rarely more than 1000 mg/dL in dogs).9 Leukocyte counts
cervical hyperesthesia.9,24 Some dogs show signs that resemble can range from 2 to more than 1000 cells/µL in both dogs and
those of cervical disc herniation. The most common ocular cats. A mixed cellular pleocytosis is generally present, with
abnormalities are chorioretinitis, papilledema, evidence of optic neutrophils or large mononuclear cells generally outnumbering
neuritis, and retinal detachment. Decreased nasal airflow may other cell types. Some dogs have a marked eosinophilic pleocy-
be present in dogs with nasal cavity cryptococcomas. Nodules, tosis. Cryptococcal yeasts are visible in most but not all animals
masses, or ulcers may involve any portion of the integument, and with cryptococcal meningitis (see Figure 62-2).9 Marked deteri-
the nose, tongue, buccal mucosa, hard palate, lips, or nail beds. oration in neurologic status occasionally occurs in animals with
CNS cryptococcosis immediately after CSF collection. Many
Diagnosis animals have intracranial mass lesions or increased CSF pres-
sure, which can increase the risk of cerebellar herniation fol-
Definitive diagnosis of cryptococcosis is usually straightforward lowing a cisternal tap. Wherever possible, attempts to make a
and based on evaluation of representative tissue specimens with diagnosis with serum antigen testing or collection of specimens
cytology, culture, and occasionally histopathology, with or with- from other anatomic sites should be considered as an alternative
out serologic detection of cryptococcal antigen in body fluids to diagnosis by CSF analysis.
(Table 62-1). Suitable specimens include nasal swabs, nasal wash-
ings, needle aspirates from mass lesions or enlarged lymph nodes, Diagnostic Imaging
bronchoalveolar lavage specimens, pleural fluid, CSF, and urine. Plain Radiography
Thoracic radiographs in dogs and cats are often normal. In
Laboratory Abnormalities some animals, interstitial to alveolar infiltrates or small nodu-
Complete Blood Count, Serum Chemistry Profile, and Urinalysis lar lesions are present. Occasionally, large pulmonary nodules,
Changes in the CBC, chemistry panel, and urinalysis in animals hilar lymphadenopathy, a mediastinal granuloma, or pleural
with cryptococcosis are generally mild and nonspecific. Mild to effusion are found.41
CHAPTER 62  Cryptococcosis 605

FIGURE 62-7  Transverse T1-weighted pre-contrast (left), T1-weighted post-contrast (center), and T2-weighted (right) MRI of the brain of an 8-year-old male neutered domestic short-
hair with cryptococcosis. There is a focal lesion in the gray matter of the right cerebral cortex at the junction of the parietal and occipital lobes (arrowhead). The lesion is T2 hyperintense, T1
hypointense, and rim-enhancing (arrow). These signal characteristics were consistent with a pseudocyst lesion.

Computed Tomography
In cats with nasal cryptococcosis, computed tomography (CT)
of the nasal cavity can reveal soft tissue and fluid opacifica-
tion of the nasal cavity or frontal sinus, peripherally contrast-
enhancing mass lesions of the nasal planum, and lysis of the
nasal bones or of the cribriform plate.11 In dogs, CT findings in
the nasal cavity include peripherally contrast-enhancing mass
lesions within the frontal sinuses, caudal nasal cavity, nasophar-
ynx, or the retrobulbar space and associated osteomyelitis (see
Figure 62-4, A). CNS abnormalities such as ventricular dila-
tion and intra-axial contrast-enhancing mass lesions have been
described in dogs using CT.9 However, MRI is more sensitive
for detection of CNS lesions in human patients,42 and the same
is probably true for dogs and cats.

Magnetic Resonance Imaging


MRI abnormalities in cats with CNS cryptococcosis consist of
single or multifocal contrast-enhancing mass lesions that tend
to be hyperintense on T2-weighted images and hypointense
on T1-weighted images, as well as mild to moderate, diffuse
or focal meningeal enhancement.9 Some cats have lesions that FIGURE 62-8  India ink stained preparation of Cryptococcus gattii yeasts that had
appear fluid-filled on T2-weighted images but have a greater been grown in culture. 1000× oil magnification. (Courtesy Greg Hodges and George
T1-weighted intensity than expected for acellular fluid (“high Thompson III, University of California, Davis.)
T2, low T1 lesions”) (Figure 62-7).9 MRI in dogs may show
contrast-enhancing mass lesions that are hyperintense on
T2-weighted images and of variable intensity on T1-weighted Microbiologic Tests
images, as well as meningeal enhancement (see Figure 62-4, B). Cytologic Examination
Ventriculomegaly and/or cribriform plate osteomyelitis may Cytologic examination can be performed on aspirates, body flu-
also be evident. ids, or impression smears of biopsy specimens. Romanowsky-
type stains, new methylene blue, and Gram stain are all
Sonographic Findings satisfactory for making a cytologic diagnosis. India ink can be
Over 80% of cats with cryptococcosis have normal abdomi- used to examine CSF for cryptococcal yeasts, which appear
nal ultrasound examinations. Cats with kidney involvement unstained and silhouetted against a black background. India ink
can have iso- to hypoechoic intraparenchymal mass lesions staining of CSF is still used routinely in the diagnosis of human
on ultrasound, with or without associated renal failure. These cryptococcal infections (Figure 62-8).42 However, lymphocytes
sometimes involve the renal pelvis. However, cats with renal and fat droplets can sometimes be confused with the organism.
involvement can also have normal abdominal ultrasound Although cytologic examination of nasal or cutaneous exu-
examinations.11 dates, aspirates, or body fluids for organisms is a rapid and
Ultrasonographic findings in dogs include intra-­abdominal sensitive test for cryptococcosis, negative test results do not
lymphadenomegaly, multifocal gastric and small intestinal thick- eliminate the possibility of cryptococcosis. Generally, organ-
ening, changes suggestive of pancreatitis, renal mass lesions, isms are found only by cytologic examination of CSF in 60% to
and/or ascites (see Figure 62-3).11,48 80% of dogs and cats.11 If a sufficient index of suspicion exists,
606 SECTION 3  Fungal and Algal Diseases

additional diagnostic tests such as histopathology, antigen test-


ing, and/or fungal culture are indicated.

Fungal Culture
Cryptococcus spp. are readily cultured from aspirates, exudate,
CSF, urine, and tissue specimens. Attempts to culture the organ-
ism are recommended whenever possible before treatment is
initiated, because isolation confirms the diagnosis, can be used
to differentiate C. gattii from C. neoformans, and permits anti-
fungal susceptibility testing for cases that subsequently fail to
respond adequately to antifungal drug treatment. Some veteri-
nary diagnostic laboratories still report all Cryptococcus iso-
lates as C. neoformans in the absence of tests to discriminate
between C. neoformans and C. gattii. If the report lists C. neo-
formans as the isolate, the laboratory must therefore be con-
sulted to determine whether C. neoformans was differentiated
FIGURE 62-9  Growth of C. neoformans (left) and C. gattii (right) on canavanine-
from C. gattii. However, whether the clinician needs to know glycine-bromothymol blue agar. Cryptococcus neoformans does not grow well in the pres-
whether the isolate is C. neoformans or C. gattii is debatable ence of canavanine and does not utilize glycine, so the medium color remains unchanged.
because at this time, species identification does not clearly influ- Cryptococcus gattii grows in the presence of l-canavanine and utilizes glycine as the sole
ence treatment recommendations or prognosis. If Cryptococcus carbon and nitrogen source, which raises the pH of the medium and causes the bromothy-
is isolated from a contaminated site such as the nasal cavity, a mol blue indicator in the medium to turn the agar a vivid cobalt blue color.
diagnosis of cryptococcosis should be supported with cytologic
or histopathologic evidence of active infection. This is because
subclinical colonization occurs in some animals. human literature it has been suggested that when the MIC rises
C. neoformans and C. gattii grow on almost all laboratory during treatment, or if the initial MIC is at least 16 µg/mL for
media, but fungal isolation media (such as Sabouraud’s dextrose fluconazole, or at least 128 µg/mL for flucytosine, failure of
agar) and incubation temperatures of 25°C or 30°C in addition antifungal drug treatment may result from drug resistance.42
to incubation at 37°C are preferred when fungi are being con-
sidered in the differential diagnosis. Antibiotics are added to the Serologic Testing
medium for isolation of Cryptococcus spp. from contaminated Latex agglutination assays for detection of cryptococcal poly-
sites, such as the nasal cavity. Colonies usually are visible in 2 to saccharide capsular antigen are widely used for diagnosis of
3 days, but sometimes incubation for up to 10 days is required. cryptococcosis in veterinary patients and are sensitive and
Unlike other dimorphic fungi, Cryptococcus grows as a yeast, specific.11,52,53 ELISA assays are also available, and the perfor-
rather than a mold, on routine fungal media and so is less likely mance of a rapid lateral-flow assay (CrAg, IMMY Diagnostics,
to represent a laboratory hazard than organisms that grow as Norman, OK) is under investigation in the authors’ labora-
molds. The organisms form white, creamy colonies. Often tories. Serologic assays detect the antigen of all C. gattii and
C. gattii colonies are more mucoid than C. neoformans colonies. C. neoformans molecular types and can be used on serum or
The organism is identified based on its microscopic morphology CSF. These can provide a rapid diagnosis when organisms
and response to biochemical tests in commercial yeast identifica- cannot be visualized or cultured. When positive, serum cryp-
tion kits (see Chapter 4). Canavanine-glycine-bromothymol blue tococcal antigen assays can be advantageous in animals with
(CGB) agar is used to differentiate between C. neoformans and neurologic signs when CSF collection is unacceptably risky or if
C. gattii.50 Unlike C. neoformans, C. gattii grows on CGB agar a diagnosis of cryptococcosis is unlikely but requires additional
and turns the media a brilliant deep blue (Figure 62-9). Fungal evidence for exclusion.
culture of the CSF is frequently positive in patients with CNS In human patients, commercial kits typically have at least
cryptococcosis and should be performed even if the organism 90% sensitivity and specificity.42 Results have been similar for
cannot be demonstrated cytologically or serologically. Culture small animal patients,11,52,53 although false negative results may
of urine specimens collected using cystocentesis is also indicated be more common in dogs than in cats.11 False positive test results
when cryptococcosis is suspected, even when obvious renal are reduced when test specimens are pretreated in the laboratory
involvement is not present and cytologic abnormalities are not with pronase (a proteinase).42 In human patients, titers below
present. Identification of molecular types for epidemiologic pur- 1:8 can represent false-positive results, although false-positive
poses requires specialized techniques such as multilocus sequence titers of up to 1:200 have rarely been detected in cats.11 These
typing or PCR fingerprinting. However, matrix-assisted laser cats had other causative etiologies identified that explained their
desorption/ionization–time of flight mass spectrometry (MALDI- clinical signs. In some cats these low positive titers may repre-
TOF) can also rapidly differentiate between Cryptococcus spe- sent subclinical colonization. Attempts to obtain cytologic or
cies as well as molecular types, and may be used in veterinary histopathologic confirmation of infection in animals with titers
clinical microbiology laboratories in the future.51 of 1:200 or less are recommended.
Most cryptococcal isolates from dogs or cats are susceptible Antigen titers are frequently extremely high (>1:65,536) in
to amphotericin B, 5-flucytosine, and azoles. Rarely, resistance cats and dogs with cryptococcosis, but even a titer of 1:2 can
to 5-flucytosine and azole antifungal drugs (especially flucon- indicate cryptococcal infection.52 Titer results from different
azole) is detected. In general, minimum inhibitory concentra- kits vary considerably; therefore, the methodology should not
tions (MICs) for fluconazole are higher than those for other be altered when monitoring the response to treatment. A good
azole drugs, and this does not imply resistance in vivo. In the prognosis is indicated by a decrease in antigen titer, whereas
CHAPTER 62  Cryptococcosis 607

FIGURE 62-10  Histopathology showing the meninges of a cat with cryptococcosis. Numerous round yeast organisms occupy the meningeal spaces. H&E stain.

a persistent titer after treatment suggests continued infection. stained with hematoxylin-eosin, Cryptococcus yeasts are faint,
Reductions in the titer typically lag behind clinical improve- eosinophilic, round to oval bodies that are surrounded by a
ment. No correlation has been found between pretreatment clear halo (the unstained capsule). The organism is more eas-
antigen titer and outcome. ily visualized with periodic acid–Schiff, methenamine silver,
Because the sensitivity of cryptococcal antigen testing is not or Fontana-Masson stain, although the capsule still does not
100%, cryptococcosis should not be ruled out on the basis of stain well with these methods. With Mayer’s mucicarmine
a negative antigen titer alone. This is especially important in stain, the cryptococcal capsule takes on a rose-red color, and
dogs, because false negatives are more common in dogs than in the organism appears pink against a blue background. Other
cats.11 False negatives are most common in animals with local- fungi with similar morphologic features do not stain with
ized ocular, CNS, or solitary cutaneous lesions. For dogs and Mayer’s mucicarmine. Some Cryptococcus strains have poorly
cats suspected to have cryptococcal meningoencephalomyelitis developed capsules. If necessary, immunohistochemistry can
that have negative serum antigen titers, CSF should be submit- be used to differentiate Cryptococcus from other fungi and
ted for cytologic examination, antigen titers, and fungal culture. some reagents can provide information on the identity of the
species and serotype present.55
Molecular Diagnosis Using the Polymerase Chain Reaction
Real-time PCR assays for Cryptococcus spp. are offered on a Cats
commercial basis by some veterinary diagnostic laboratories. In cats that die or are euthanized as a result of cryptococco-
These assays can confirm the presence of Cryptococcus DNA in sis, histopathology of lesions within the nasal cavity may reveal
tissues or body fluids when other means fail. However, because granulomatous rhinitis. CNS cryptococcosis consists of either
a diagnosis of cryptococcosis is usually apparent based on the meningitis41 or meningoencephalomyelitis, with single or mul-
methods described earlier, PCR is currently not regarded as a tiple cerebral granulomas (Figure 62-10). Extension of infection
routinely useful clinical tool. Because Cryptococcus DNA has from the meninges along perforating arteries into the Virchow-
been detected using PCR assays in small intestinal contents Robin spaces can result in cystic collections of yeast organisms
from healthy dogs and dogs with chronic enteropathies,54 posi- known as gelatinous pseudocysts.9 Ocular lesions include pyo-
tive results on intestinal biopsies may not reflect ­cryptococcosis. granulomatous to granulomatous chorioretinitis, optic neuritis,
Similarly, because subclinical nasal cavity colonization can uveitis, endophthalmitis, panophthalmitis, and retinal separa-
occur, positive results on nasal biopsies may not represent inva- tion. Other affected organs include skin and subcutaneous tis-
sive disease. Because false positives also have the potential to sues, kidneys, and lymph nodes. Renal granulomas have been
occur as a result of contamination in the PCR laboratory, a found in some cats with disseminated disease. Granulomas can
PCR diagnosis of cryptococcosis should always be supported by also occasionally be found in the spleen, adrenal glands, thyroid
cytologic or histopathologic evidence of infection and/or culture glands, and liver.
of Cryptococcus spp. from a normally sterile site.
Dogs
Pathologic Findings Dogs that die or are euthanized because of cryptococcosis often
Lesions associated with cryptococcosis vary from gelatinous have CNS infection, ocular involvement, or widely disseminated
masses that consist almost exclusively of organisms to well- disease. The lesions consist of granulomatous or pyogranulo-
ordered granulomas or pyogranulomas that typically occur in matous meningoencephalitis, sometimes with pseudocyst for-
patients with improved cell-mediated immune (CMI) responses. mation; neuritis of the optic, facial, or vestibular nerves; and
The gelatinous appearance is a reflection of the large amount pyogranulomatous or granulomatous chorioretinitis. Inflam-
of capsular polysaccharide present in lesions. In sections matory lesions that contain Cryptococcus yeasts may also be
608 SECTION 3  Fungal and Algal Diseases

present in the lungs, nasal cavity, kidneys, pancreas, liver, myo- is used alone, it is used chiefly to improve the efficacy of other
cardium, gastrointestinal tract, and spleen. Less commonly, antifungal drugs. Lipid-complexed amphotericin B prepara-
involvement of the peritoneum, thyroid, tongue, prostate, tions are not necessarily more effective but are less nephro-
pleura, joints, mediastinum, urinary bladder, and adrenal gland toxic, which may translate into greater efficacy because higher
has been detected at necropsy. doses can be used, although at a substantially increased cost. In
human patients, higher doses of amphotericin B more effectively
Treatment and Prognosis sterilize the CSF. The reader is referred to Chapter 9 for dosing
protocols for various formulations of amphotericin B. Treat-
Treatment recommendations for dogs and cats with cryptococ- ment with 5-flucytosine (or fluconazole) and amphotericin B is
cosis have been based on evidence-based studies performed in continued for 2 to 4 weeks (ideally 4 weeks) or until azotemia
human patients and clinical experiences; evidence-based studies develops, after which treatment is continued with azole mono-
are lacking for veterinary patients. The most common problems therapy. Additional courses of amphotericin B may be required
encountered are the high cost of therapy, requirement for mul- to maintain clinical remission in some cats. Although controlled
tiple hospital visits, and the need for regular medication of pets trials have not been performed, some cats with CNS cryptococ-
for a protracted period. Immunocompromised animals (which cosis have shown apparent response to addition of terbinafine
may include purebred dogs with no obvious underlying immu- to the treatment regimen, when other drugs failed to resolve
nodeficiency) are especially likely to have persistent or progres- clinical signs.57
sive infection in the face of treatment. In many affected animals,
treatment must be continued for years or the life of the animal. Dogs
Surgical debulking could be considered before treatment for If affordable to the owner, dogs with disseminated disease or
large cryptococcomas to improve drug penetration. However, CNS involvement should be treated initially with deoxycho-
even large cryptococcomas can resolve with medical treatment late or lipid-complexed amphotericin B and fluconazole. Dogs
alone, and surgery should only be performed if lesions can be treated with flucytosine frequently develop toxic epidermal
easily and safely resected. The reader is referred to Chapter 9 for necrolysis, typically 10 to 14 days after starting therapy, which
detailed information on antifungal drugs. precludes treatment with flucytosine in this species.58 Some
dogs with CNS involvement respond to azole monotherapy,
Cats although residual neurologic signs may persist.9 Other potential
Cats with localized cutaneous disease can sometimes be treated outcomes include incomplete recovery, with persistent active
successfully with azole monotherapy. The initial drug of choice infection despite therapy, or relapse after prolonged clinical
is fluconazole (10 mg/kg PO q12h), because of its good penetra- remission. In one dog, surgical debridement of an extradural
tion of the brain, eye, and urinary tract; low cost; and minimal spinal cord granuloma combined with fluconazole treatment
adverse effects. controlled spinal cord infection.59 Again, azoles are fungistatic
Treatment should be continued until there is complete lesion and rely on a CMI response and phagocytosis to remove crypto-
resolution and the cryptococcal antigen titer becomes negative. coccal yeasts from the host, which may be lacking in some dogs
A four- to fivefold reduction in the antigen titer suggests an with cryptococcosis. Treatment of a dog with intestinal crypto-
adequate response. In cats with C. gattii molecular type VGI or coccosis with terbinafine was associated with long-term disease
C. neoformans infections, this typically takes 2 to 12 months resolution when treatment with azoles, surgical debulking, and
(median, 4 months),56 although longer periods of therapy may amphotericin B failed.48
be required for some cats. For recovered animals that achieve
an antigen titer of 0, the titer can be monitored every 3 to Other Antifungal Drugs
6 months, so that any recurrence is diagnosed early. However, Other antifungal drugs that are effective for treatment of cryp-
in the authors’ experience, many cats with C. gattii molecular tococcosis include voriconazole and posaconazole, although
type VGII and VGIII infections in North America fail to respond these can be unaffordable for many clients. Voriconazole pene-
completely to fluconazole treatment, antigen titers persist at high trates the CNS and so is potentially advantageous for treatment
levels, and relapse occurs after an initial treatment response. of cryptococcal meningoencephalitis. However, virtually all cats
In some of these cats, treatment with itraconazole (5-10 mg/ and some dogs do not tolerate voriconazole treatment because
kg PO q24h [capsules] or 3-5 mg/kg PO q24h [solution]) or of gastrointestinal, neurologic, and cardiac adverse effects. In
ketoconazole can lead to clinical remission. Liver enzyme activi- humans, posaconazole is well tolerated, and it has been used to
ties should be monitored monthly during treatment with azole treat other fungal infections without complication in cats (see
antifungal drugs to monitor for hepatotoxicity (see Chapter 9). Chapter 9), so it could be an option for cats with refractory dis-
Amphotericin B is the most effective anticryptococcal ease. Unpredictable serum levels have led to increased interest in
agent. Penetration of the CNS and vitreous is poor, but clini- therapeutic serum level determination in humans.
cal responses still occur in cats and dogs with CNS and ocular
cryptococcosis, presumably because the blood-brain barrier is Supportive Care
compromised. A combination of amphotericin B and 5-flucy- Because many cats and dogs with CNS involvement deterio-
tosine (250 mg PO q8h) is considered the optimal therapy for rate clinically during initial treatment with antifungal drugs
cats (and humans, but not dogs) with CNS cryptococcosis, or as a result of the inflammatory response to dying yeasts, anti-­
for those that fail to respond to azole drugs. Fluconazole can inflammatory doses of short-acting glucocorticoids may be nec-
be substituted for 5-flucytosine if 5-flucytosine is unaffordable. essary to support animals with CNS cryptococcosis for the first
However, 5-flucytosine shows synergy when combined with few days to weeks of treatment and may increase survival rate
amphotericin B and effectively penetrates the blood-brain bar- at 10 days after initiation of treatment.9 Deterioration of neu-
rier. Because resistance can develop rapidly when 5-flucytosine rologic status in the first few days of treatment is not a negative
CHAPTER 62  Cryptococcosis 609

prognostic indicator if the animal can be supported through this to less than 50 to 100 cells/µL correlates well with the risk of
phase. Supportive care with IV fluid therapy, tube feeding, and infection in HIV-infected humans.42 No vaccines are available.
antiseizure medications may be necessary. Rarely, dogs or cats
require supplemental oxygen because of respiratory involvement. Prevention
Prognosis Prevention of cryptococcosis may be possible through avoid-
In general, cats show significant clinical improvement within ance of exposure to ecologic niches of the organism, such as
1 to 2 weeks of starting therapy. In studies from Australia, pigeon feces. Nevertheless, cryptococcosis occurs in animals
around 75% of cats were treated successfully.5,56 As many as housed exclusively indoors, and widespread exposure and
60% of cats may be cured after an initial course of therapy, recovery likely occurs in endemic areas.
although some cats that show an initial favorable response to
therapy can relapse, sometimes as long as 10 years after therapy Public Health Aspects
is discontinued.56 In one study, the median survival time of cats
with CNS cryptococcosis was only 13 days (range, 0 to 4050 Cryptococcosis is an important opportunistic fungal infection
days), but a median survival time was never reached if cats sur- of people with AIDS, in particular those who are not treated
vived at least 3 days after diagnosis, so supportive care during with highly active antiretroviral therapy. Human infection is
this phase may be critical.9 acquired from the environment and not by direct contact with
The prognosis is more guarded for dogs. Six of 11 dogs with infected animals, although intense exposure to avian excrement
cryptococcosis in Australia were treated successfully.56 In a has been associated with some cryptococcosis cases in HIV-
study from the western United States9 and another from south- infected humans.62 Malignancy, chemotherapy, immunosup-
western Australia,5 the success rate was lower, around 30%. pressive drug therapy, diabetes mellitus, splenectomy, cirrhosis,
The median survival time for dogs with CNS cryptococcosis or sarcoidosis can also predispose to the disease. Severe menin-
was 7 days (range, 0 to 3680 days).9 gitis and disseminated infections can develop in immunodefi-
In human patients with cryptococcosis, negative prognostic cient patients.
indicators include underlying immunosuppressive disease or The organism does not aerosolize from sites of tissue infec-
drugs, abnormal mental state, high organism burden at presen- tion, so the disease cannot spread among people or animals.
tation (as defined by large numbers of yeasts in the CSF and/or The major public health significance of infected pets is that
high antigen titers), and a poor anti-inflammatory response in cats may act as a sentinel for infection of human beings,
the CSF (<20 cells/µL).60 The presence of CNS disease appears because of their susceptibility to infection. In the diagnostic
to negatively affect outcome in both cats and dogs.56,61 In dogs laboratory, culture of Cryptococcus is not a significant health
and cats with CNS involvement, the presence of altered mental hazard, because only the yeast form is grown routinely, and
status at first evaluation was associated with increased mortality this form does not aerosolize from media. During handling
(P = 0.03).9 of infected tissues or cultures, precautions should be taken to
prevent inadvertent cutaneous inoculation of the organism.
Immunity and Vaccination Veterinarians should consider patient sedation for procedures
that involve fine-needle aspiration in order to avoid inadver-
Cryptococcosis can occur in apparently immunocompetent or tent needle-stick injuries, which have caused localized cuta-
in immunodeficient individuals, although those with deficiencies neous infection in the human health care setting.63 Because
of cell-mediated immunity are most commonly infected. Pro- sporotrichosis is a major differential diagnosis for cats with
duction of Th1 type cytokines such as Il-2, TNF-α, and IFN-γ cutaneous lesions, gloves should always be worn and hand
and a granulomatous inflammatory response are important for hygiene performed when handling cats with suspicious lesions
elimination of infection, and decline in the CD4+ T cell count (see Chapter 64).

CASE EXAMPLE acid and enrofloxacin for 7 days), and he was treated with
subcutaneous fluids (lactated Ringer’s solution, 150 mL,
once). Over the following 5 days his clinical signs lessened,
Signalment: “Simon,” an 8-year-old male neutered domestic but then anorexia developed. Physical examination 2 days
shorthair cat from northern California after the onset of anorexia revealed a rectal temperature
History: Simon was evaluated by the University of California, of 102.5°F (39.2°C) and weight loss (3.7 kg at the previous
Davis, Veterinary Medical Teaching Hospital (VMTH) for visit to 3.2 kg). A CBC and biochemistry panel showed mild
a 5-week history of progressive weakness, inappetence, lymphopenia (1157 cells/µL, reference range 1500-7000
and weight loss. Initially the owners noted that he cells/µL). Treatment with metronidazole (39 mg/kg PO q24h)
was reluctant to jump and had a low tail carriage, and and prednisone (3 mg/kg PO q24h) was instituted. Three
intermittent episodes of head shaking and sneezing were days before he was brought to the VMTH, he began panting
also noted. He also had episodes of clenching the furniture and vocalizing. The metronidazole was discontinued and the
with his claws. Three weeks before he was brought to the prednisone dose was reduced to 1.5 mg/kg PO q24h. The
VMTH, Simon was seen at a local veterinary clinic where a following day, the owners noted that Simon’s eyelids were
physical examination was unremarkable. Broad-spectrum twitching, and he urinated and defecated on himself in the
antimicrobial drugs were prescribed (amoxicillin-clavulanic litter box. Clindamycin (6 mg/kg PO q12h) was prescribed

Continued
610 SECTION 3  Fungal and Algal Diseases

for suspected toxoplasmosis, and Simon was referred for Total protein 7.5 g/dL (6.6-8.4 g/dL)
further evaluation. Albumin 3.8 g/dL (2.2-4.6 g/dL)
Simon had no other previous medical or surgical history Globulin 3.7 g/dL (2.8-5.4 g/dL)
apart from being neutered as a kitten. He had access to ALT 37 U/L (27-101 U/L)
the outdoors and there were no other cats in the house- AST 28 U/L (17-58 U/L)
hold. There had been no changes in his thirst or urina- ALP 8 U/L (14-71 U/L)
tion, no vomiting or diarrhea, and he was last vaccinated GGT < 3 U/L (0-4 U/L)
at 5 years of age. He had killed a large rat ­approximately Cholesterol 177 mg/dL (89-258 mg/dL)
5 weeks earlier and had not been himself since. Total bilirubin < 0.1 mg/dL (0-0.2 mg/dL).
Physical Examination: Urinalysis: SGr 1.018; pH 7.0; 25 mg/dL protein; no bilirubin,
Body Weight: 2.9 kg. hemoprotein, glucose; rare WBC/HPF, 0 RBC/HPF, few
General: Obtunded but responsive, estimated as 5% amorphous crystals, few lipid droplets.
dehydrated. T = 100.2°F (37.9°C), HR = 200 beats/min, RR = ELISA for FeLV Antigen and FIV Antibody: Negative.
60 breaths/min, CRT <2 s. Latex Agglutination for Cryptococcus Antigen: Positive at
Eyes, Ears, Nose, and Throat: Bilateral mydriasis and 1:32,768.
absent pupillary light reflexes were noted. There was no Imaging Findings:
evidence of nasal discharge and nasal airflow was normal. Plain Thoracic and Abdominal Radiographs: The only
Mucous membranes were pink and slightly tacky. Fundic abnormality present was mild lateral spondylosis of the
examination was within normal limits. caudal lumbar spine.
Musculoskeletal: Body condition score 4/9, ambulatory. The Abdominal Ultrasonography: The liver was mildly enlarged
cat had a slow gait with a short stride. and contained multiple small hyperechoic foci. In the
Respiratory: Tachypnea was present with a normal respiratory kidneys, mild cortical hyperechogenicity and decreased
effort. Normal breath sounds were present on auscultation. corticomedullary distinction were present.
Cardiovascular, Gastrointestinal, Genitourinary and Magnetic Resonance Imaging (Brain): Precontrast images
Lymph Nodes: No clinically significant abnormalities were showed a well-defined, round T2-hyperintense, T1-
detected. The urinary bladder was small. hypointense lesion in the gray matter of the right cerebral
Neurologic Examination: The cat was mildly obtunded, cortex at the junction of the parietal and occipital lobes
walked with a crouched stance and behaved as if nonvisual. (see Figure 62-7). Postcontrast images showed moderate
Bilateral mydriasis and absent menace and pupillary light contrast enhancement of this lesion. There was mild contrast
reflexes were confirmed. Intermittent horizontal nystagmus enhancement of the meninges ventral to the right preoptic
with the fast phase to the left was present. Cranial nerve area. There was moderate, focal contrast enhancement of
examination was otherwise normal. Placing and hopping the gray matter at the lateral aspect of the right pyriform
reactions were delayed in the left pelvic limb. There was a lobe. There was contrast enhancement of the medial
decreased left patellar reflex, but other segmental reflexes aspect of both pyriform lobes adjacent to the hypophysis.
were intact. There was no apparent pain on spinal palpation, Impressions: Multifocal contrast-enhancing lesions with T2
but the cat appeared painful when the tail was raised. hyperintensity and a cystic lesion in the right cerebral cortex.
Neuroanatomic location: Multifocal CNS disease. Cytologic Findings: CSF analysis (cisternal): Protein
Laboratory Findings: concentration 29 mg/dL (reference range, <25 mg/dL). The
CBC: specimen had a moderate increase in cellularity with a low
HCT 27.2% (30%-50%) number of erythrocytes. Numerous thick-capsuled yeast
MCV 48.3 fL (42-53 fL) organisms with frequent narrow budding forms were noted,
MCHC 29.8 g/dL (30-33.5 g/dL) consistent with Cryptococcus spp. Nucleated cells consisted
Reticulocytes 8400 cells/µL (7000-60,000 cells/µL) of nondegenerate neutrophils admixed with low numbers
WBC 12,360 cells/µL (4500-14,000 cells/µL) of variably vacuolated macrophages and small lymphocytes.
Neutrophils 11,000 cells/µL (2000-9000 cells/µL) The differential cell count was 83% neutrophils, 7% small
Lymphocytes 828 cells/µL (1000-7000 cells/µL) mononuclear cells, and 10% large mononuclear cells.
Monocytes 445 cells/µL (50-600 cells/µL) Microbiologic Testing: Fungal culture (CSF): Cryptococcus
Eosinophils 87 cells/µL (150-1100 cells/µL) gattii. Multilocus sequence typing revealed the organism
Basophils 0 cells/µL (0-50 cells/µL) belonged to C. gattii molecular type VGIIa.
Platelets 357,000/µL (180,000-500,000 platelets/µL). Diagnosis: Cryptococcus gattii meningoencephalitis.
Serum Chemistry Profile: Treatment: Simon was treated with deoxycholate
Sodium 150 mmol/L (151-158 mmol/L) amphotericin B (0.25 mg/kg in 30 mL 5% dextrose IV over
Potassium 4.3 mmol/L (3.6-4.9 mmol/L) 30 minutes on Monday, Wednesday, and Friday for five
Chloride 115 mmol/L (117-126 mmol/L) treatments). Before and after this treatment, 0.9% NaCl
Bicarbonate 19 mmol/L (15-21 mmol/L) was administered at twice maintenance rate and kidney
Phosphorus 5.4 mg/dL (3.2-6.3 mg/dL) analytes were monitored. Flucytosine (62.5 mg PO q8h)
Calcium 10 mg/dL (9.0-10.9 mg/dl) and prednisone (2.5 mg PO q24h) were also administered.
BUN 21 mg/dL (18-33 mg/dL) An esophagostomy tube was placed for enteral nutrition
Creatinine 1.2 mg/dL (1.1-2.2 mg/dL) because of persistent inappetence. By day 7, Simon
Glucose 136 mg/dL (63-118 mg/dL) appeared more alert, pupillary light reflexes and menace
CHAPTER 62  Cryptococcosis 611

responses were present but weak, and nystagmus had additional month, the antigen titer was 1:1024, a serum
resolved. He was discharged from the hospital on day 12, chemistry profile was unremarkable, and the owners felt
with instructions to continue treatment with fluconazole that Simon was a normal cat. The prednisone treatment
(50 mg PO q12h) and prednisone (2.5 mg PO q48h), in was discontinued thereafter. A year later, the antigen titer
addition to esophagostomy tube feeding. On day 24, he was 1:20. At a recheck examination 2 years later the cat was
was reevaluated for lethargy of several days’ duration. neurologically normal. The owner declined further antigen
Physical examination showed resolution of cranial nerve testing and chose to continue treatment with itraconazole.
abnormalities, but delayed hopping reactions in all Comments: This cat had CNS cryptococcosis caused by
limbs were present. He was treated with six additional Cryptococcus gattii molecular type VGIIa, a hypervirulent
amphotericin B treatments over 2 weeks. He began molecular type that emerged in British Columbia and the
eating, and the esophageal feeding tube was removed Pacific Northwest. The treatment plan was limited by client
on day 40. Three months after diagnosis, his neurologic financial concerns, which meant that the initial course of
signs had almost completely resolved with the exception amphotericin B and flucytosine treatment was short, and a
of occasional tremors, persistent mild placing reaction second course of amphotericin B was required. Subsequently,
deficits, and lumbosacral pain. A cryptococcal antigen treatment with fluconazole was associated with limited
titer was positive at 1:65,536. The fluconazole was replaced improvement, but a change to itraconazole was followed by
with itraconazole solution (3 mg/kg PO q12h). After an dramatic clinical improvement and reduction in antigen titers.

SUGGESTED READINGS 11. Trivedi SR, Sykes JE, Cannon MS, et al. Clinical features and epide-
miology of cryptococcosis in cats and dogs in California: 93 cases
Datta K, Bartlett KH, Baer R, et al. Spread of Cryptococcus gattii into (1988-2010). J Am Vet Med Assoc. 2011;239:357-369.
Pacific Northwest region of the United States. Emerg Infect Dis. 12. Kano R, Kitagawat M, Oota S, et al. First case of feline systemic
2009;15:1185-1191. Cryptococcus albidus infection. Med Mycol. 2008;46:75-77.
Kronstad JW, Attarian R, Cadieux B, et  al. Expanding fungal patho- 13. Labrecque O, Sylvestre D, Messier S. Systemic Cryptococcus
genesis: Cryptococcus breaks out of the opportunistic box. Nat Rev albidus infection in a Doberman Pinscher. J Vet Diagn Invest.
Microbiol. 2011;9:193-203. 2005;17:598-600.
Trivedi SR, Malik R, Meyer W, et  al. Feline cryptococcosis: impact 14. Johnson LB, Bradley SF, Kauffman CA. Fungaemia due to Crypto-
of current research on clinical management. J Feline Med Surg. coccus laurentii and a review of non-neoformans cryptococcaemia.
2011;13:163-172. Mycoses. 1998;41:277-280.
15. Kano R, Hosaka S, Hasegawa A. First isolation of Cryptococcus
REFERENCES magnus from a cat. Mycopathologia. 2004;157:263-264.
1. Knoke M, Schwesinger G. One hundred years ago: the history of 16. Bernardo FM, Martins HM, Martins ML. A survey of
cryptococcosis in Greifswald. Medical mycology in the nineteenth mycotic otitis externa of dogs in Lisbon. Rev Iberoam Micol.
century. Mycoses. 1994;37:229-233. 1998;15:163-165.
2. Stephen C, Lester S, Black W, et al. Multispecies outbreak of cryp- 17. Kronstad JW, Attarian R, Cadieux B, et  al. Expanding fungal
tococcosis on southern Vancouver Island, British Columbia. Can pathogenesis: Cryptococcus breaks out of the opportunistic box.
Vet J. 2002;43:792-794. Nat Rev Microbiol. 2011;9:193-203.
3. Lester SJ, Kowalewich NJ, Bartlett KH, et  al. Clinicopathologic 18. Wickes BL, Mayorga ME, Edman U, et  al. Dimorphism
features of an unusual outbreak of cryptococcosis in dogs, cats, and haploid fruiting in Cryptococcus neoformans: associa-
ferrets, and a bird: 38 cases (January to July 2003). J Am Vet Med tion with the alpha-mating type. Proc Natl Acad Sci U S A.
Assoc. 2004;225:1716-1722. 1996;93:7327-7331.
4. Rotstein DS, West K, Levine G, et al. Cryptococcus gattii VGI in 19. Hull CM, Heitman J. Genetics of Cryptococcus neoformans. Annu
a spinner dolphin (Stenella longirostris) from Hawaii. J Zoo Wildl Rev Genet. 2002;36:557-615.
Med. 2010;41:181-183. 20. Velagapudi R, Hsueh YP, Geunes-Boyer S, et  al. Spores as infec-
5. McGill S, Malik R, Saul N, et al. Cryptococcosis in domestic ani- tious propagules of Cryptococcus neoformans. Infect Immun.
mals in Western Australia: a retrospective study from 1995-2006. 2009;77:4345-4355.
Med Mycol. 2009;47:625-639. 21. Ngamskulrungroj P, Gilgado F, Faganello J, et al. Genetic diversity
6. Datta K, Bartlett KH, Baer R, et al. Spread of Cryptococcus gattii of the Cryptococcus species complex suggests that Cryptococcus
into Pacific Northwest region of the United States. Emerg Infect gattii deserves to have varieties. PLoS One. 2009;4:e5862.
Dis. 2009;15:1185-1191. 22. Malik R, Wigney DI, Muir DB, et al. Cryptococcosis in cats: clini-
7. Krockenberger MB, Canfield PJ, Barnes J, et  al. Cryptococ- cal and mycological assessment of 29 cases and evaluation of treat-
cus neoformans var. gattii in the koala (Phascolarctos cinereus): ment using orally administered fluconazole. J Med Vet Mycol.
serological evidence for subclinical cryptococcosis. Med Mycol. 1992;30:133-144.
2002;40:273-282. 23. O’Brien CR, Krockenberger MB, Wigney DI, et  al. Retrospective
8. Malik R, Alderton B, Finlaison D, et al. Cryptococcosis in ferrets: a study of feline and canine cryptococcosis in Australia from 1981 to
diverse spectrum of clinical disease. Aust Vet J. 2002;80:749-755. 2001: 195 cases. Med Mycol. 2004;42:449-460.
9. Sykes JE, Sturges BK, Cannon MS, et al. Clinical signs, imaging fea- 24. Berthelin CF, Bailey CS, Kass PH, et al. Cryptococcosis of the ner-
tures, neuropathology, and outcome in cats and dogs with central vous system in dogs: part 1. Epidemiologic, clinical, and neuro-
nervous system cryptococcosis from California. J Vet Intern Med. pathologic features. Prog Vet Neurol. 1994;5:88-97.
2010;24:1427-1438. 25. Malik R, Dill-Macky E, Martin P, et  al. Cryptococcosis in dogs:
10. Malik R, Krockenberger MB, Cross G, et al. Avian cryptococcosis. a retrospective study of 20 consecutive cases. J Med Vet Mycol.
Med Mycol. 2003;41:115-124. 1995;33:291-297.
612 SECTION 3  Fungal and Algal Diseases

26. Jacobs GJ, Medleau L, Calvert C, et al. Cryptococcal infection in 45. Georgi A, Schneemann M, Tintelnot K, et  al. Cryptococcus gat-
cats: factors influencing treatment outcome, and results of sequen- tii meningoencephalitis in an immunocompetent person 13 months
tial serum antigen titers in 35 cats. J Vet Intern Med. 1997;11:1-4. after exposure. Infection. 2009;37:370-373.
27. Kano R, Fujino Y, Takamoto N, et al. PCR detection of the Cryp- 46. MacDougall L, Fyfe M. Emergence of Cryptococcus gattii in a
tococcus neoformans CAPS9 gene from a biopsy specimen from a novel environment provides clues to its incubation period. J Clin
case of feline cryptococcosis. J Vet Diagn Invest. 2001;13:439-442. Microbiol. 2006;44:1851-1852.
28. Barrs VR, Martin P, Nicoll RG, et  al. Pulmonary cryptococcosis 47. Beatty JA, Barrs VR, Swinney GR, et al. Peripheral vestibular dis-
and Capillaria aerophila infection in an FIV-positive cat. Aust Vet J. ease associated with cryptococcosis in three cats. J Feline Med Surg.
2000;78:154-158. 2000;2:29-34.
29. Nielsen K, De Obaldia AL, Heitman J. Cryptococcus neoformans 48. Olsen GL, Deitz KL, Flaherty HA, et al. Use of terbinafine in the
mates on pigeon guano: implications for the realized ecological treatment protocol of intestinal Cryptococcus neoformans in a dog.
niche and globalization. Eukaryot Cell. 2007;6:949-959. J Am Anim Hosp Assoc. 2012;48:216-220.
30. Lazera MS, Pires FD, Camillo-Coura L, et  al. Natural habitat of 49. Malik R, Hunt GB, Bellenger CR, et al. Intra-abdominal cryptococ-
Cryptococcus neoformans var. neoformans in decaying wood form- cosis in two dogs. J Small Anim Pract. 1999;40:387-391.
ing hollows in living trees. J Med Vet Mycol. 1996;34:127-131. 50. Kwon-Chung KJ, Polacheck I, Bennett JE. Improved diagnos-
31. Trivedi SR, Malik R, Meyer W, et al. Feline cryptococcosis: impact tic medium for separation of Cryptococcus neoformans var.
of current research on clinical management. J Feline Med Surg. neoformans (serotypes A and D) and Cryptococcus neofor-
2011;13:163-172. mans var. gattii (serotypes B and C). J Clin Microbiol. 1982;15:
32. Viviani MA, Cogliati M, Esposto MC, et  al. Molecular analy- 535-537.
sis of 311 Cryptococcus neoformans isolates from a 30-month 51. Firacative C, Trilles L, Meyer W. MALDI-TOF MS enables the
ECMM survey of cryptococcosis in Europe. FEMS Yeast Res. rapid identification of the major molecular types within the Cryp-
2006;6:614-619. tococcus neoformans/Cryptococcus gattii species complex. PLoS
33. Sorrell TC. Cryptococcus neoformans variety gattii. Med Mycol. One. 2012;7:e37566.
2001;39:155-168. 52. Malik R, McPetrie R, Wigney DI, et al. A latex cryptococcal anti-
34. Marr KA. Cryptococcus gattii as an important fungal patho- gen agglutination test for diagnosis and monitoring of therapy for
gen of western North America. Expert Rev Anti Infect Ther. cryptococcosis. Aust Vet J. 1996;74:358-364.
2012;10:637-643. 53. Medleau L, Marks MA, Brown J, et  al. Clinical evaluation of a
35. Bartlett KH, Cheng PY, Duncan C, et  al. A decade of experi- cryptococcal antigen latex agglutination test for diagnosis of cryptococ-
ence: Cryptococcus gattii in British Columbia. Mycopathologia. cosis in cats. J Am Vet Med Assoc. 1990;196:1470-1473.
2012;173:311-319. 54. Suchodolski JS, Morris EK, Allenspach K, et al. Prevalence and iden-
36. MacDougall L, Kidd SE, Galanis E, et  al. Spread of Cryptococ- tification of fungal DNA in the small intestine of healthy dogs and dogs
cus gattii in British Columbia, Canada, and detection in the Pacific with chronic enteropathies. Vet Microbiol. 2008;132:379-388.
Northwest. USA. Emerg Infect Dis. 2007;13:42-50. 55. Krockenberger MB, Canfield PJ, Kozel TR, et al. An immunohis-
37. Duncan CG, Stephen C, Campbell J. Evaluation of risk factors tochemical method that differentiates Cryptococcus neoformans
for Cryptococcus gattii infection in dogs and cats. J Am Vet Med varieties and serotypes in formalin-fixed paraffin-embedded tissues.
Assoc. 2006;228:377-382. Med Mycol. 2001;39:523-533.
38. Mitchell DH, Sorrell TC, Allworth AM, et al. Cryptococcal disease 56. O’Brien CR, Krockenberger MB, Martin P, et al. Long-term out-
of the CNS in immunocompetent hosts: influence of cryptococcal come of therapy for 59 cats and 11 dogs with cryptococcosis. Aust
variety on clinical manifestations and outcome. Clin Infect Dis. Vet J. 2006;84:384-392.
1995;20:611-616. 57. Sykes JE. Unpublished observations, 2012.
39. Campbell LT, Currie BJ, Krockenberger M, et  al. Clonality and 58. Malik R, Medeiros C, Wigney DI, et  al. Suspected drug erup-
recombination in genetically differentiated subgroups of Crypto- tion in seven dogs during administration of flucytosine. Aust Vet J.
coccus gattii. Eukaryot Cell. 2005;4:1403-1409. 1996;74:285-288.
40. Speed B, Dunt D. Clinical and host differences between infections 59. Kerwin SC, McCarthy RJ, VanSteenhouse JL, et  al. Cervical spi-
with the two varieties of Cryptococcus neoformans. Clin Infect Dis. nal cord compression caused by cryptococcosis in a dog: successful
1995;21:28-34: discussion 35–26. treatment with surgery and fluconazole. J Am Anim Hosp Assoc.
41. Chaturvedi S, Dyavaiah M, Larsen RA, et  al. Cryptococcus gat- 1998;34:523-526.
tii in AIDS patients, southern California. Emerg Infect Dis. 60. Bicanic T, Harrison TS. Cryptococcal meningitis. Br Med Bull.
2005;11:1686-1692. 2004;72:99-118.
42. Perfect JR. Cryptococcus neoformans. In: Mandell GL, Bennett JE, 61. Duncan C, Stephen C, Campbell J. Clinical characteristics and pre-
Dolin R, eds. Principles and Practice of Infectious Diseases. 7th ed. dictors of mortality for Cryptococcus gattii infection in dogs and cats
Philadelphia, PA: Elsevier; 2010:3287-3303. of southwestern British Columbia. Can Vet J. 2006;47:993-998.
43. Perfect JR. The triple threat of cryptococcosis: it’s the body site, the 62. Fessel WJ. Cryptococcal meningitis after unusual exposures to
strain, and/or the host. MBio. 2012;3. birds. N Engl J Med. 1993;328:1354-1355.
44. Perfect JR. Cryptococcus neoformans: a sugar-coated killer 63. Casadevall A, Mukherjee J, Yuan R, et al. Management of injuries
with designer genes. FEMS Immunol Med Microbiol. 2005;45: caused by Cryptococcus neoformans–contaminated needles. Clin
395-404. Infect Dis. 1994;19:951-953.
CHAPTER 63

Coccidioidomycosis
Jane E. Sykes

In the environment, Coccidioides spp. are distributed in


Overview of Coccidioidomycosis the lower Sonoran life zone, regions characterized by semiarid
First Described: 1892 (by the medical student Alejandro Posa- to arid soils, low elevations above sea level, and hot summers.
das), in a person from Buenos Aires, Argentina, where it Worldwide, Coccidioides spp. are found in the southwest-
was initially mistaken for a protozoan (Coccidia)1 ern United States, Mexico, and parts of Central and South
America, specifically Colombia, Guatemala, Honduras, Ven-
Cause: Coccidioides immitis and Coccidioides posadasii (asco-
ezuela, northeastern Brazil, Argentina, Paraguay, and Bolivia
mycetes, no known teleomorph)
(Figure 63-2). In the southwestern United States, highly
Affected Hosts: Primarily humans and dogs, but also a variety endemic regions are the south-central valley of California,
of other animals that include cats, domestic farm animal especially Bakersfield (“valley fever”) and Arizona, especially
species, and a variety of wildlife hosts Tucson and Phoenix.2 Infections also occur in non-endemic
Geographic Distribution: Southwestern United States, Mex- regions in dogs and humans that have a travel history to
ico, and parts of Central and South America (northeastern endemic regions.
Brazil, Argentina, Paraguay, and Bolivia) Two species of Coccidioides have been identified, C. posa-
dasii and C. immitis. These appear to cause similar clinical
Mode of Transmission: Inhalation of arthroconidia from the
manifestations and have similar antifungal drug suscepti-
environment; rarely cutaneous inoculation
bilities, but further studies are required.3 The geographic
Major Clinical Signs: Cough, fever, inappetence, weight loss, range of C. immitis is largely limited to the central valley of
tachypnea, increased respiratory effort, lameness, subcu- California, whereas C. posadasii is found elsewhere.4 How-
taneous masses or draining skin lesions, lymphadenopa- ever, there may be some overlap in the distribution of these
thy, neurologic signs, ascites due to right-sided heart organisms.
failure (pericarditis), ocular lesions (uveitis, chorioretinitis, Infection of animals and humans with Coccidioides spp.
endophthalmitis) often follows a cycle of moist conditions (required for growth
Differential Diagnoses: Neoplasia, other deep mycoses, myco- of the organism in the soil), a dry period, then soil disrup-
bacteriosis, actinomycosis or nocardiosis, toxoplasmosis tion, such as may occur with heavy rainfall, earthquakes, dust
or neosporosis, systemic immune-mediated disease storms, prolonged droughts, or construction.5-7 Dogs of any
breed, age, or sex may be affected, although large-breed, young
Human Health Significance: Direct transmission from ani- adult dogs predominate in case series.8,9 In dogs seen at the
mals to humans does not usually occur, although sharps University of California Veterinary Medical Teaching Hospi-
injuries or bite wounds from affected animals may be a tal, boxers, poodles, pointers, Labrador and golden retrievers,
source of human infection. Dogs are a sentinel for human German shepherds, Australian shepherds, Dalmatians, Scottish
exposure. terriers, dachshunds, and beagles are overrepresented when
compared with the hospital patient population.10 Risk factors
for infection in dogs from Arizona include being housed out-
doors during the day rather than indoors, roaming areas more
Etiology and Epidemiology than 1 acre, and walking in the desert.11 Latent infections may
also occur in dogs, and these can reactivate after treatment
Coccidioides spp. are dimorphic, soil-borne fungi that exist with immunosuppressive drugs, such as corticosteroids and
in the environment as a mycelium and in tissues (at 37°C) as chemotherapeutic agents, including after previous recovery
peculiar structures known as spherules. The mycelium consists from coccidioidomycosis.
of chains of haploid, multinucleated, barrel-shaped arthroco- Although not as prevalent as in dogs, coccidioidomycosis
nidia (or arthrospores) that alternate with smaller, thin-walled, also occurs in cats.12 In a study of 48 cats from Arizona, cats
nonviable cells. The arthroconidia remain viable in the soil for ranged in age from 1 to 15 years (median, 5 years) and there was
long periods. They ultimately fragment from each other and are no obvious sex or breed predisposition.12 One cat was infected
subsequently aerosolized and inhaled by animal hosts (Figure with FIV, and all cats tested were FeLV negative. Interestingly,
63-1). This can be followed by the development of coccidioi- a search of the author’s hospital and pathology database from
domycosis, a localized pulmonary disease or a serious multisys- 1990 to 2012 revealed not a single cat with coccidioidomycosis,
temic disease if dissemination occurs in the face of inadequate which might suggest cats are less susceptible to C. immitis infec-
immune responses. tion than to C. posadasii infection.

613
614 SECTION 3  Fungal and Algal Diseases

FIGURE 63-1  Life cycle of Coccidioides spp. Hyphae fragment into arthroconidia, which are inhaled and form spherules in tissues. The spherules rupture and release endospores, which
form new spherules. In some dogs, the organism disseminates from the lungs to a variety of other tissues, but especially local lymph nodes, bone, skin, the pericardium, the eye, and the
central nervous system.

Clinical Features
Signs and Their Pathogenesis
The clinical signs of coccidioidomycosis depend on factors
such as the infectious dose and the immune status of the host.
Arthroconidia are inhaled and are phagocytosed by alveolar
macrophages. They then enlarge into a spherule, which mea-
sures 8 to 100 µm in diameter. Hundreds of endospores then
develop within the spherule, which are released when the mature
spherule ruptures. These attract neutrophils, which leads to a
pyogranulomatous inflammatory response. Each released endo-
spore that escapes immune destruction then enlarges into a
new spherule, and the cycle continues. Endospores can revert
to mycelial growth when removed from the site of infection. In
hosts that are unable to mount an effective immune response,
endospores disseminate by lymphatics to tracheobronchial
lymph nodes and hematogenously to a variety of body sites.
Inoculation coccidioidomycosis can rarely occur, and generally
results in lesions that are localized to the site of the wound. In
one report, it followed removal of a foxtail from a wound.13
Most infections in endemic areas are subclinical.14 Develop-
ment of clinical signs often follows a subacute to chronic course,
Highly endemic
with variable and often mild systemic signs such as intermittent
Moderately endemic
fever, lethargy, inappetence, and weight loss.8,15 Many dogs oth-
Mildly endemic
erwise appear healthy, with occasional periods of inappetence.8
Suspected endemic
Clinical signs may be present for days to years before veteri-
nary attention is sought.9 Signs of respiratory tract involvement
include a harsh cough, increased respiratory effort, tachypnea
and/or exercise intolerance.9,15 Cough may be chronic and is
FIGURE 63-2  Geographic distribution of Coccidioides spp. worldwide. The inset
sometimes associated with gagging or retching; occasionally it
shows the distribution of Coccidioides species in the southwestern United States.
results from bronchial compression by enlarged tracheobron-
chial lymph nodes. Rarely, diffuse pneumonia follows a fulmi-
nant course, with signs that resemble acute bacterial pneumonia
or septic shock, as reported in immunocompromised human
patients.2
CHAPTER 63  Coccidioidomycosis 615

pericardial coccidioidomycosis may have abdominal distention


with a palpable fluid wave, tachycardia, tachypnea, jugular
pulses, muffled breath and heart sounds due to pleural effu-
sion, mucosal pallor, prolonged capillary refill time, and weak
pulses.20 Neurologic signs, when present, include obtundation,
apparent blindness, ataxia, nystagmus, spinal pain, circling,
and tetraparesis. The most common ocular abnormality is uve-
itis, but keratitis, conjunctivitis, granulomatous chorioretinitis,
optic neuritis, retinal detachment, and endophthalmitis with
secondary glaucoma have also been described.9,18,19
Of 48 cats with coccidioidomycosis, fever was present in
31%. Skin lesions were present in more than 50% of 48 affected
cats and included draining skin lesions, subcutaneous granulo-
mas, and abscesses.12 Tachypnea or increased respiratory effort
was present in 25% of affected cats, and lameness in fewer than
20% of cats. Ocular abnormalities were present in 13% of cats.
Conjunctival masses, periorbital swellings, chorioretinitis, reti-
nal detachment, endophthalmitis, and/or anterior uveitis have
been described.21,22 A small percentage of cats have neurologic
signs such as hyperesthesia, posterior paresis, seizures, and
FIGURE 63-3  Draining skin lesion on the left elbow of a 5-year-old shepherd mix ataxia. Dissemination to abdominal organs such as the spleen
with disseminated coccidioidomycosis. Radiography revealed a mixed productive and can occur.23
destructive lesion of the distal humerus and proximal ulna. Productive lesions of the sixth
and seventh ribs were also present. Arthrocentesis revealed suppurative inflammation Diagnosis
with no growth on culture. Diagnosis was ultimately made by biopsy of the synovium and
serology. The Coccidioides titer was 1:64. Coccidioidomycosis is usually suspected based on a history of
travel to or residence in an endemic region, and consistent clini-
A smaller percentage of infected dogs develop disseminated cal signs and radiographic findings. Because the disease is not
infections. Sites of dissemination often include osteoarticular prevalent in some endemic regions (such as coastal southern
sites, the central nervous system (CNS), skin, peripheral lymph California), early diagnosis requires a high index of suspicion
nodes, eyes, testes, prostate, and the pericardium.9,10 Rarely, in these regions. The diagnosis can be confirmed with cytologic
sites such as the liver, spleen, gastrointestinal tract, or urinary examination of aspirates or body fluids, serologic assays that
system are affected.8 Sometimes dissemination to only one ana- detect antibodies to Coccidioides spp., histopathology, and/or
tomic site is evident; in other dogs multiple sites are involved. A fungal culture (Table 63-1).
history of respiratory signs may be absent.8 Bone involvement
may manifest as lameness and/or one or more firm swellings Laboratory Abnormalities
associated with the appendicular or axial skeleton. Dogs with Complete Blood Count
skin involvement may have subcutaneous masses ulcerated CBC findings in dogs with coccidioidomycosis are nonspecific
skin lesions that drain serosanguineous fluid (Figure 63-3).8 and resemble those of other deep mycoses. Most affected dogs
Skin lesions may overlie sites of osteomyelitis. Neurologic signs have mild, normocytic, normochromic, nonregenerative anemia
result from meningoencephalitis and include obtundation, and mild leukocytosis. Leukocytosis results from a neutrophilia,
blindness, nystagmus, absent menace reflexes, diminished gag occasionally with a left shift and evidence of neutrophil toxic-
responses, ataxia, abnormal placing reactions, pacing, circling, ity.9,20 A monocytosis and lymphopenia may also be present,
cervical pain, tetraparesis, and seizures.9,16,17 Ocular mani- but in contrast to human infections, eosinophilia is not typically
festations of coccidioidomycosis include chorioretinitis, uve- present.
itis, optic neuritis, and endophthalmitis.9,18,19 Involvement of
abdominal organs such as the liver and gastrointestinal tract Serum Biochemical Tests
can lead to inappetence, vomiting, and/or diarrhea.8 Pericardi- Almost all dogs with coccidioidomycosis have hypoalbumin-
tis may lead to signs of right-sided cardiac failure, with ascites emia, and approximately half of affected dogs and cats have
and pleural effusion.8 hyperglobulinemia (up to 7.8 mg/dL in one study).9,12 Hypoal-
buminemia is usually mild to moderate, but rarely may be more
Physical Examination Findings severe (<1.5 g/dL). Uncommonly, mild hypercalcemia is present.
Fever is present in up to two thirds of dogs with coccidioidomy- Increased activity of serum ALP may be present in some dogs,
cosis (range, 103°F to 106°F) on physical examination. Other possibly as a result of bone involvement.
common findings in dogs are lethargy, weakness, thin body
condition or profound muscle atrophy, tachypnea, increased Urinalysis
respiratory effort, inducible or spontaneous cough, and/or The urinalysis is often normal, but significant proteinuria (urine
increased lung sounds.9 Lameness and firm, often painful bony protein-to-creatinine ratios as high as 9.5, reference range <1)
swellings with associated muscle atrophy may be present in has been reported in some dogs with coccidioidomycosis.9 It
dogs with osteomyelitis. Less often, focal or (very rarely) gen- is not clear whether proteinuria of this magnitude results from
eralized peripheral lymphadenomegaly, subcutaneous masses or glomerulonephritis secondary to chronic immune stimulation or
draining skin lesions, or swollen joints are present. Dogs with another comorbidity.
616 SECTION 3  Fungal and Algal Diseases

TABLE 63-1
Diagnostic Assays Currently Available for Coccidioidomycosis in Dogs and Cats
Assay Specimen Type Target Performance
Cytologic Aspirates of affected tissues, Coccidioides spherules Low sensitivity and organisms may be difficult to
examination body fluids, impression and/or endospores accurately identify in some specimens. Immature
smears of skin lesions spherules may be mistaken for other fungi or cel-
lular aggregates or debris.
Fungal culture Aspirates or biopsies of affect- Coccidioides spp. Generally sensitive and specific, but false negatives
ed tissues, body fluids such are possible if specimen size is low. Can require
as CSF or pleural effusion, several weeks’ incubation. A significant labora-
transtracheal or bronchoal- tory health hazard.
veolar lavage specimens
Antibody serology Serum, CSF IgG or IgM antibodies Sensitivity is close to 100%. Quantification of an
(gel immunodiffu- to Coccidioides spp. IgG antibody titer is possible. Titers decline with
sion) successful treatment. In hyperendemic areas,
titers as high as 1:16 have the potential to exist
in subclinically infected or recovered dogs, so
positive serology should be supported with other
diagnostic tests for coccidioidomycosis.
Serology for Serum, urine Coccidioides spp. Low sensitivity (≤20%); false positives can occur in
Coccidioides galactomannan antigen dogs with histoplasmosis or blastomycosis. Not
antigen recommended.
Real-time PCR Biopsies, body fluids, respi- Coccidioides DNA Not yet validated in adequate numbers of dogs or
assays ratory lavage specimens, cats with coccidioidomycosis; usefulness requires
aspirates further evaluation.
Histopathology Biopsy specimens from Coccidioides spp. Limited sensitivity in dogs; submission of multiple
affected tissues, such spherules biopsies is recommended when possible. Often
as bone biopsies only one or two spherules are visualized. Special
stains such as periodic acid–Schiff and silver
stains facilitate organism detection.

Cerebrospinal Fluid Analysis Radiographs of affected bones in both dogs and cats with
CSF analysis in dogs with CNS coccidioidomycosis usually reveals Coccidioides osteomyelitis reveal local periosteal proliferation
an increase in protein concentration (generally <300 mg/dL, as well as osteolysis and soft tissue swelling. Lesions can resem-
reference range <25 mg/dL) and a mild to moderate increase in ble osteosarcomas (Figure 63-5).8,12
the nucleated cell count (usually <50 cells/µL, reference range
<2 cells/µL).9 The differential cell count may show mixed, pre- Sonographic Findings
dominantly lymphocytic, mixed mononuclear, or neutrophilic Abdominal ultrasound examination of dogs with coccidioido-
pleocytosis. Organisms are generally not observed. mycosis is typically unremarkable. Mild hepatosplenomegaly
and abdominal lymphadenomegaly have been described in
Diagnostic Imaging a small proportion of dogs.9 Ascites, hepatomegaly, and dis-
Plain Radiography tended hepatic veins may be evident in dogs with pericarditis.20
Thoracic radiographs in dogs with disseminated coccidioi- Echocardiography in dogs with Coccidioides spp. pericardi-
domycosis are often unremarkable. The most common radio- tis can reveal thickening and mass lesions of the pericardium,
graphic finding in dogs with pulmonary coccidioidomycosis adherence of the pericardium to the epicardium, pericardial
is mild to severe hilar lymphadenomegaly (Figure 63-4, A), and pleural effusion, and evidence of cardiac tamponade (see
which was present in 10 of 19 dogs with coccidioidomycosis Figure 86-3).20
from California.9 Hilar lymphadenopathy can also be present
in affected cats.12 Although hilar lymphadenopathy can be the Advanced Imaging
only finding, most dogs with hilar lymphadenopathy also have Advanced imaging findings in dogs with coccidioidomyco-
pulmonary infiltrates, which are usually mild to moderate and sis have not been extensively reported in the literature. Focal,
interstitial. Nodular interstitial, interstitial-alveolar, bronchoin- contrast-enhancing intra-parenchymal mass lesions in the brain
terstitial infiltrates, and/or sternal lymphadenomegaly can also or osteomyelitis of the skull may be identified on computed
be present (Figure 63-4, B and C).9,15 Dogs with pericarditis can tomography (CT) of the head.24 Thoracic CT findings include
have cardiomegaly and pleural effusion, with hepatomegaly and contrast-enhancing miliary nodular pulmonary lesions; single or
decreased abdominal detail.20 multifocal larger nodules or masses; tracheobronchial, sternal,
CHAPTER 63  Coccidioidomycosis 617

or mediastinal lymphadenopathy; focal regions of alveolar or


peribronchial infiltrates; lobar consolidation; pleural or medi-
astinal thickening; and, rarely, pleural effusion (Figure 63-6).
MRI of the brain in dogs with CNS coccidioidomyco-
sis may show focal hyperintense lesions within the brain on
T2-weighted images (Figure 63-7). These are typically isointense
on T1-weighted images and enhance with contrast. Other find-
ings include ventricular dilation and/or meningeal enhancement.

Microbiologic Tests
Cytologic Examination
Cytologic examination of aspirates of affected tissues, pleu-
ral effusion, or respiratory lavage specimens typically reveals
granulomatous or pyogranulomatous inflammation, sometimes
with rare multinucleated giant cells, scattered eosinophils, and/
A or a reactive lymphocyte population. However, occasionally a
suppurative inflammatory response predominates. Spherules
may be visualized as round, deeply basophilic, double-walled,
slightly crinkled structures that range in diameter from 8 to
70 µm. Spherules are generally low in number, frequently asso-
ciated with cellular aggregates, but often they are not evident
or easily identified. Usually their internal structure is not easily
discernible, but occasionally endospores are seen in association
with ruptured spherules (Figure 63-8). The endospores are 2 to 5
µm in diameter, are surrounded by a thin, nonstaining halo, and
have small, round to oval, densely aggregated, eccentric nuclei.
Endospores may be found extracellularly or within phagocytes.
In one report, only endospores were visualized in a fine-needle
aspirate from a bone lesion; intact spherules were not seen.13
Because cytology is relatively insensitive for diagnosis of
coccidioidomycosis when compared with other deep mycoses,
serologic testing for coccidioidomycosis should always be con-
B sidered when cytology of tissue aspirates, body fluids, or lung
washes reveals pyogranulomatous, suppurative, or granuloma-
tous inflammation but organisms are not seen, and exposure to
Coccidioides is possible based on the history.

Serologic Diagnosis
Serologic assays that detect antibodies to Coccidioides spp.
are important diagnostic tests for coccidioidomycosis because
organism detection assays have low sensitivity. Most diagnostic
laboratories screen serum specimens with qualitative gel immu-
nodiffusion (ID) assays for IgG or IgM antibodies (see Chapter
2). Detection of IgM (tube precipitin) or IgG (complement fixat-
ing) antibodies is reported, depending on the antigen prepara-
tion used in the assay. In dogs, IgM is detectable within 2 to
5 weeks of infection, and IgG appears after 8 to 12 weeks.10
Quantification of an IgG antibody response can then be per-
formed by quantitative ID, whereby serial dilutions of patient
serum are subjected to gel ID.14 In human patients, the com-
plement fixation test is often used to quantify IgG antibody,
but frequent false-positive results have been described with
this method in dogs as a result of anticomplementary proper-
C ties of canine serum.25 The complement fixation test is positive
FIGURE 63-4  Thoracic radiographic patterns in coccidioidomycosis. A, Lateral thoracic in most cats with coccidioidomycosis; titers have ranged from
radiograph from a 7-year-old intact male English pointer that shows several tracheobronchial 1:2 to 1:128.12
lymphadenomegaly and focal pulmonary infiltrates. B, Lateral thoracic radiograph from a False-negative serologic test results are extremely rare in dogs
3-year-old intact female Rottweiler with cough, anorexia, and respiratory distress. There with coccidioidomycosis tested at the author’s institution,9,20
are diffuse nodular and peribronchial infiltrates throughout the pulmonary parenchyma but they have been described in dogs from Arizona.26,27 Anti-
and possible hilar lymphadenopathy. Necropsy revealed necrogranulomatous interstitial
body titers usually range from 1:2 to 1:256; the median titer
pneumonia with intralesional spherules. The spleen, liver, and kidney were also involved. 
C, Dorsoventral thoracic radiograph from a 2-year-old female spayed Labrador retriever with was 1:32 in one study.9 There is no clear correlation between
cough and inappetence. There is an alveolar pattern in the left cranial lung lobe. the magnitude of the IgG titer and the severity of illness in dogs.
618 SECTION 3  Fungal and Algal Diseases

A B C
FIGURE 63-5  Osteomyelitis caused by Coccidioides spp. A, Left tarsus of a 4-year-old female spayed Dalmatian that was evaluated for signs of right-sided congestive heart failure due
to pericarditis. Severe osteolysis is present. B, Right humerus of a 4-year-old female spayed shepherd mix. There is a mixed productive and lytic lesion of the distal diaphysis of the right
humerus. C, Lesion in (B) at necropsy. (Courtesy University of California, Davis, Veterinary Anatomic Pathology Service.)

FIGURE 63-6  Computed tomography scan of the thorax of a 2-year-old intact male
Labrador retriever with coccidioidomycosis. There are infiltrates in the caudal aspect of the
right cranial lung lobe that extend from the mediastinum to the pleural surface. The walls
of the right cranial bronchus and associated airways are thickened. Bronchial lymphadeno- B
megaly was also identified. FIGURE 63-7  MRI scan of the brain of a 7-year-old male neutered shih tzu with
disseminated coccidioidomycosis. A, A small focus of increased signal intensity on
T2-weighting is present in the left lateral aspect of the brainstem (arrowhead). Ventricular
dilation is also present (arrow). B, T1-weighted image after administration of gadolinium.
The lesion in the brainstem is contrast enhancing.
CHAPTER 63  Coccidioidomycosis 619

data suggest that this assay has unacceptable sensitivity (≤20%)


for diagnosis of coccidioidomycosis in dogs when serum, urine, or
both specimens are tested, and false-positive reactions occur in a
small proportion of dogs with blastomycosis or histoplasmosis.29

Fungal Culture
Coccidioides spp. can be isolated from clinical specimens on rou-
tine fungal media in the laboratory, but a negative culture result
does not rule out coccidioidomycosis. Growth of a gray-white,
downy mold that is tan to brown when visualized through the
bottom of the plate usually appears after incubation for several
days. Isolates can be identified based on their light microscopic
appearance, with or without molecular methods. Culture of
Coccidioides spp. is a serious laboratory health hazard and
should only be performed if necessary and by suitably equipped
laboratories. The clinician should warn the laboratory of the
possibility of dimorphic fungal infection if suspected.

Molecular Diagnosis Using the Polymerase Chain Reaction


A Real-time PCR assays have been used for rapid detection of
Coccidioides spp. in respiratory specimens or CSF from affected
humans, although sensitivity has been limited.2,30 PCR assays
have also been used in epidemiologic studies to identify major
growth sites of Coccidioides spp. in soil.31 PCR assays have not
yet been widely applied to the diagnosis of coccidioidomycosis
in dogs and cats.

Pathologic Findings
Gross pathologic findings in dogs with coccidioidomycosis
include mottling and consolidation of the pulmonary paren-
chyma; single or multiple pale masses, nodular lesions, or
emphysematous bullae within the lungs (Figure 63-9); enlarged
and firm tracheobronchial, mediastinal, or sternal lymph nodes;
plaque-like lesions on the pleura; and pleural and pericardial
effusion. The interior of masses or nodules may be firm, caseous,
or liquefied. The pericardium and epicardium may be thickened
with mass lesions and adhesions between the pericardium and
epicardium.20 Ascites may also be present in dogs with pericar-
ditis. Dogs with disseminated disease may have diffuse muscle
atrophy, one or more enlarged peripheral lymph nodes, irregu-
B lar bony swellings with associated draining skin lesions, and/or
FIGURE 63-8  Cytologic appearance of Coccidioides spp. spherules. A, Spherules often evidence of arthritis and synovitis. Meningoencephalitis may be
appear as deeply basophilic, crinkled, intact, or ruptured ball-like structures (white arrow). evident as mass lesions within the brain parenchyma, ventricu-
B, Sometimes, a refractile spherule wall is appreciated (arrow) and endospores are visual- lar dilation secondary to obstruction of CSF flow by inflam-
ized as granular material within the spherule. matory lesions, regional discoloration within the brain, and/
or discoloration and thickening of the meninges. Rarely, pros-
tatic enlargement, hepatic congestion and friability, multifocal
Dogs with active infection can have titers as low as 1:2, and nodular lesions within the myocardium or other parenchymal
healthy dogs in hyperendemic areas in Arizona can have titers organs, and ocular lesions are present (see Physical Examination
as high as 1:16.14 As a result, positive titers of 1:16 or lower Findings for a description of ocular lesions).
must be interpreted in light of clinicopathologic abnormalities, Histopathology of affected tissues reveals large numbers of
and in hyperendemic areas, other diagnostic tests (such as neutrophils and macrophages and sometimes marked fibrosis,
­culture, cytology or histopathology) should be used to ­confirm with occasional endosporulating or non-endosporulating spher-
the diagnosis if possible. Latex agglutination and ELISA (EIA) ules. Multinucleated giant cells may be present and can contain
assays for detection of anti-Coccidioides antibody have also spherules.26 Spherules have a thick wall, and endospores may
been developed, but these are not widely used in veterinary be visualized in mature spherules (Figure 63-10). Often only
medicine, and their use has been associated with a significant one or two spherules are visualized, and in some cases they are
number of false-positive reactions in humans.2,25,28 not found at all. Use of special stains such as periodic acid–
A serologic assay that detects Coccidioides antigen in urine is Schiff and Gomori’s methenamine silver can enhance organism
available for diagnosis of coccidioidomycosis in human patients, identification within lesions. The presence of large numbers of
such as in immunocompromised patients who lack the ability to spherules has been described in some affected cats.23,32 Chronic,
produce significant quantities of antibody. However, preliminary diffuse glomerulonephritis was present in 3 of 13 dogs with
620 SECTION 3  Fungal and Algal Diseases

FIGURE 63-9  Lungs of the dog in Figure 63-4, B. Multiple pale nodules are present throughout the lung parenchyma.

for dogs with CNS involvement, because of its superior penetra-


tion of the CNS. The dose of fluconazole used by the author is
5 mg/kg PO q12h. Itraconazole (5 mg/kg PO q24h) is preferred
for animals with bone involvement and may be effective in dogs
that fail to respond to treatment with fluconazole. In human
patients, itraconazole demonstrated a greater response rate in
a blinded comparison with fluconazole.33 Compounded formu-
lations of itraconazole should be avoided. Small dogs are best
treated with itraconazole solution (10 mg/mL), which has higher
bioavailability than the capsules (see Chapter 9). If capsules are
used, they should be administered with food. If clinical signs do
not improve with azole treatment, therapeutic drug monitoring
could be considered to ensure that serum drug concentrations
are adequate. Periodic monitoring of liver enzymes is recom-
mended for animals treated with azole antifungals.
Treatment with deoxycholate or lipid-complexed amphoteri-
cin B is recommended either alone or in combination with an
FIGURE 63-10  Histopathology of the tarsus of the dog in Figure 63-5, A. A single azole for dogs with refractory or severe disease (see Chapter 9
mature spherule is surrounded by a pyogranulomatous inflammatory response. The spher- for dosing protocols). However, studies that compare ampho-
ule contains many endospores. tericin B to azole monotherapy in dogs have not been performed.
Voriconazole and posaconazole have been used to treat refrac-
tory coccidioidomycosis in human patients, but they are not con-
coccidioidomycosis that had full necropsy examinations at the sidered first-line drugs.2 Their use for treatment of affected dogs
author’s hospital; an additional dog with renal lesions had mul- or cats is limited by expense. Voriconazole could be considered
tiple renal pyogranulomas with intralesional spherules.24 for treatment of refractory meningoencephalitis if client finances
permit its use, because it has excellent penetration of the CNS.
Treatment and Prognosis
Surgical Management
Successful treatment of coccidioidomycosis is limited by the high Ultimately, some dogs with severe and persistent osteomyelitis
cost of treatment and the often advanced nature of the disease require amputation to control the infection, and enucleation
by the time it is diagnosed in some dogs. Treatment consists of a may be required for dogs with endophthalmitis. Successful treat-
combination of antifungal drug therapy and supportive care; in ment of Coccidioides pericarditis has been reported after sub-
some cases, surgery is required. Usually a minimum of 6 months total pericardiectomy and epicardial excision, which should be
of treatment is required. performed in conjunction with antifungal drug therapy. Refer-
ral to a specialist surgeon with experience with the technique is
Antimicrobial Treatment strongly recommended. The perioperative mortality rate in one
Animals with coccidioidomycosis can respond well to azole study was reported as 24%, and the 2-year survival rate after
monotherapy with fluconazole or itraconazole. Ketoconazole discharge was 82%.20
has been used successfully to treat dogs and cats in the past but
is no longer the drug of first choice owing to its lower activity Supportive Care
and increased rate of adverse effects. Fluconazole is the least Other treatments that may be required for dogs with severe
expensive azole and may be a better choice than itraconazole pulmonary coccidioidomycosis are similar to those described
CHAPTER 63  Coccidioidomycosis 621

for blastomycosis and include oxygen supplementation, nebu- Public Health Aspects
lization and coupage, drainage of thoracic effusions, nutri-
tional support, anti-inflammatory drugs, and intravenous fluid Coccidioidomycosis is a serious infection in human patients and
therapy. Concurrent treatment with glucocorticoids and anti- has similar clinical manifestations to those described in dogs.
seizure medications may be necessary to reduce the severity of The incidence of human coccidioidomycosis has increased in
neurologic signs in dogs with CNS coccidioidomycosis. Topical Arizona in the last decade, possibly as a result of population
prednisolone acetate solution and antiglaucoma agents may be growth, immigration of immunologically naïve people, and con-
required for dogs with ocular lesions. struction. Construction and agricultural workers, archeologists,
and excavators are at high risk for coccidioidomycosis.2 African
Prognosis American and Filipino patients, as well as patients with AIDS
The prognosis for resolution for coccidioidomycosis depends and women in the third trimester of pregnancy, are at highest
on the severity of infection and the extent of dissemination. The risk for disseminated disease, which occurs in approximately
duration of treatment ranges from 6 months to many years, 0.5% of infected humans.
and in some cases, lifelong treatment is needed. IgG antibody Because of their susceptibility to infection, dogs have been
titers decrease with successful treatment, which in general used as a sentinel for human exposure to environmental sources
should be continued until lesions resolve and the titer is 1:2 or of Coccidioides spp. in seroprevalence studies.34 Direct trans-
lower. Residual radiographic opacities may be present in some mission between infected veterinary patients and humans has
dogs as a result of scar tissue formation. Dogs with localized not been reported. However, fatal coccidioidomycosis occurred
pulmonary infection have the best prognosis; the prognosis in a veterinary resident after necropsy of a horse with dissemi-
appears to be poor for dogs with CNS involvement. Dogs with nated disease.35 Inoculation coccidioidomycosis was reported
Coccidioides osteomyelitis may require lifelong treatment with in a veterinary technician from Arizona after a bite from a cat
itraconazole. that was later diagnosed with disseminated disease.23 Inocula-
tion coccidioidomycosis also has the potential to occur if sharps
Immunity and Vaccination injuries occur during aspiration of affected tissues, surgical pro-
cedures, or necropsy examinations. Proper sedation or anesthe-
Immunity to Coccidioides spp. depends primarily on cell- sia should always be used if coccidioidomycosis is suspected in
mediated immunity, specifically T lymphocytes. No vaccine is dogs or cats seen at veterinary clinics and the potential for a
currently available commercially for prevention of the disease. sharps injury exists. If accidental inoculation occurs, immediate
attention should be sought from a physician. Prolonged fluco-
Prevention nazole prophylaxis is generally recommended with monitoring
of Coccidioides serology and liver function tests. Bandaging
Prevention of coccidioidomycosis requires avoidance of hyper- of skin lesions should be avoided. Necropsies should be per-
endemic areas (especially parts of Arizona and south-central formed without delay with suitable protective clothing, and the
California), which may not be possible. In endemic areas, dogs carcasses of animals that die from coccidioidomycosis should
should be kept away from sites of soil disturbance (such as con- immediately be incinerated, because conversion of the organism
struction sites) and housed indoors during dust storms; digging to a mycelial form can occur after death of an animal. For the
should be discouraged. Housing dogs indoors during the day in same reason, burial is not recommended because of the poten-
endemic regions may reduce the chance of infection. Neverthe- tial for contamination of the environment. Fluconazole prophy-
less, it should be remembered that clinical infections develop in laxis is also indicated after accidental laboratory exposure to
only a minority of exposed dogs. inocula.36

CASE EXAMPLE so a long-acting cephalosporin was administered (cefovecin,


8 mg/kg SC once). Two days later, treatment with prednisone
(0.3 mg/kg PO q12h) was initiated, which was followed
Signalment: “Jenny”, a 2-year-old female spayed Labrador by mild improvement in the clinical signs. Jenny’s owner
retriever from Oxnard in southwestern California then sought a second opinion on day 20 after onset of
History: Jenny was evaluated for a 2-month history of clinical signs, and Coccidioides serology was performed.
cough, inappetence, and lethargy. For the first 3 days The titer was positive for IgG at 1:64. The prednisone
of illness, the owner noted only an intermittent harsh, was discontinued, and treatment was initiated with
nonproductive cough. On day 4, the dog became lethargic fluconazole (6.7 mg/kg PO q12h). However, after
and inappetent, so she was taken to a local veterinary clinic. 1 month of treatment, thoracic radiographic abnormalities
Thoracic radiographs revealed alveolar infiltrates in the left were unchanged. The fluconazole was substituted with
cranial lung lobe. A diagnosis of bacterial pneumonia was a compounded itraconazole formulation (5 mg/kg PO
suspected, so Jenny was treated with enrofloxacin (6 mg/ q12h). However, after an additional 2 weeks of treatment,
kg PO q24h), amoxicillin-clavulanic acid (12.5 mg/kg PO there had been no further improvement in the cough,
q12h), and metronidazole (8 mg/kg PO q12h) for 2 weeks. so the owner was referred to the University of California,
After the first week of treatment, there was no improvement, Davis.

Continued
622 SECTION 3  Fungal and Algal Diseases

According to the owner, Jenny had a poor appetite for dog food Treatment: The itraconazole capsules prescribed at the
but was eating home-cooked chicken, rice, and vegetables. previous veterinary clinic were examined and found to
Her activity level had been low. The cough occurred many contain a white powder rather than granules. Treatment was
times throughout the day and was unrelated to exercise and changed to brand-name itraconazole (Sporanox), because
eating. Occasionally she had paroxysms of cough with a ter- compounded formulations can be unstable. Instructions
minal retch. Other medical history included canine parvovi- were given to administer the itraconazole (5 mg/kg PO
rus infection when she was 8 weeks old and “kennel cough” q24h) with food to improve absorption. Aspirin was also
when she was 9 months old, which resolved after 10 days. prescribed (12 mg/kg PO q12h). One month later Jenny’s
The owners lived in a semirural area just north of Los Angeles. cough and activity level had improved considerably. A CBC
The only travel had been to Atascadero, further north on the was within normal limits, and a biochemistry panel showed
California coast. She was the only animal in the household. hypoalbuminemia (3.3 g/dL, reference range 3.4-4.3 g/dL)
There had been ongoing construction around the house and hyperglobulinemia (3.4 g/dL, reference range 1.7-
to replace a septic system with a sewer, which began 4 to 6 3.1 g/dL). Liver enzyme activities were within reference
weeks before the onset of clinical signs. ranges. Thoracic radiographs showed resolution of
Physical Examination: hilar lymphadenopathy with persistence of the alveolar
General: Quiet but alert and responsive, hydrated. T = 102.6°F infiltrates in the left cranial lung lobe. The IgG antibody
(39.2°C), HR = 100 beats/min, RR = 24 breaths/min, CRT = 1 s. titer to Coccidioides was 1:64. Three months later, the cough
Eyes, Ears, Nose, and Throat: No abnormalities were noted. had resolved completely, and after 6 months of treatment,
A dilated fundoscopic examination was unremarkable. there was complete resolution of thoracic radiographic
Musculoskeletal: Body condition score was 4/9. The dog was abnormalities and the titer was 1:32. At this point, the owner
ambulatory with no lameness or muscle atrophy. switched to a generic formulation of itraconazole because
Respiratory: Harsh breath sounds were present in all lung of financial concerns. After an additional 3 and 6 months of
fields. A harsh cough with a terminal retch was readily treatment, the IgG titer was 1:16 and 1:8, respectively, with
elicited on tracheal palpation. no further clinical signs. Treatment was continued for an
Cardiovascular, Gastrointestinal, Genitourinary, and Lymph additional 3 months (i.e., total of 15 months of treatment),
Nodes: No clinically significant abnormalities were detected. and the dog remains alive and apparently healthy at the
Imaging Findings: Thoracic radiographs were compared to time of writing.
the two-view thoracic study performed 2 weeks previously, Comments: This is an interesting case of pulmonary
which was available for review. The initial radiographs showed coccidioidomycosis that occurred outside the hyperendemic
a dense alveolar pattern throughout the left cranial lung central valley region in California. Exposure may have
lobe and increased opacity in the region of the hilar lymph resulted from soil disturbance as a result of sewer work.
nodes with splaying of the mainstem bronchi (see Figure Because of the low suspicion for the disease, the disease
63-4, C). These abnormalities had improved slightly in the was mistaken for bacterial pneumonia. The owner had
10-day recheck interim, but alveolar infiltrates were still limited financial resources, so an extensive respiratory work-
present throughout the left cranial lung lobe. There was a up (including respiratory lavage with cytology and culture)
persistent, mild, diffuse pulmonary interstitial pattern in the was declined. When Coccidioides serology was positive,
remaining pulmonary parenchyma, which was most severe fluconazole was prescribed, but there was no improvement.
in the caudodorsal lung fields. This was considered excessive The decision to treat with compounded itraconazole
for the age of the patient. The cardiovascular structures were was made because of client financial limitations, but
within normal limits. serum drug levels with this treatment were undetectable.
Microbiologic Testing: Coccidioides serology: positive Fortunately, disease ultimately resolved with a change to a
by qualitative gel immunodiffusion for complement- noncompounded formulation, and there was progressive
fixing antibody (IgG), negative for IgM. Quantitative decline in serial IgG titers (which were always performed by
immunodiffusion revealed an IgG titer of 1:128. the same laboratory). Whether treatment beyond complete
Serum itraconazole concentration: <0.31 µg/mL (undetectable). resolution of the radiographic abnormalities was necessary
Diagnosis: Pulmonary coccidioidomycosis. is unknown.

SUGGESTED READINGS REFERENCES


Greene RT, Troy GC. Coccidioidomycosis in 48 cats: a retrospective 1. Hirschmann JV. The early history of coccidioidomycosis: 1892-
study (1984-1993). J Vet Intern Med. 1995;9:86-91. 1945. Clin Infect Dis. 2007;44:1202-1207.
Johnson LR, Herrgesell EJ, Davidson AP, et al. Clinical, clinicopatho- 2. Thompson 3rd GR. Pulmonary coccidioidomycosis. Semin Respir
logic, and radiographic findings in dogs with coccidioidomycosis: 24 Crit Care Med. 2011;32:754-763.
cases (1995-2000). J Am Vet Med Assoc. 2003;222:461-466. 3. Ramani R, Chaturvedi V. Antifungal susceptibility profiles of Coc-
Shubitz LE, Butkiewicz CD, Dial SM, et al. Incidence of Coccidioides cidioides immitis and Coccidioides posadasii from endemic and
infection among dogs residing in a region in which the organism is non-endemic areas. Mycopathologia. 2007;163:315-319.
endemic. J Am Vet Med Assoc. 2005;226:1846-1850. 4. Fisher MC, Koenig GL, White TJ, et al. Molecular and phenotypic
description of Coccidioides posadasii sp. nov., previously recog-
nized as the non-California population of Coccidioides immitis.
Mycologia. 2002;94:73-84.
CHAPTER 63  Coccidioidomycosis 623

5. Flynn NM, Hoeprich PD, Kawachi MM, et  al. An unusual 22. Tofflemire K, Betbeze C. Three cases of feline ocular coccidioido-
outbreak of windborne coccidioidomycosis. N Engl J Med. mycosis: presentation, clinical features, diagnosis, and treatment.
1979;301:358-361. Vet Ophthalmol. 2010;13:166-172.
6. Schneider E, Hajjeh RA, Spiegel RA, et  al. A coccidioidomycosis 23. Gaidici A, Saubolle MA. Transmission of coccidioidomycosis to a
outbreak following the Northridge, Calif, earthquake. JAMA. human via a cat bite. J Clin Microbiol. 2009;47:505-506.
1997;277:904-908. 24. Sykes JE. Unpublished observations, 2012.
7. Park BJ, Sigel K, Vaz V, et al. An epidemic of coccidioidomycosis in 25. Gershman N, Villaba W, Neel K. Detection of canine IgG and IgM
Arizona associated with climatic changes, 1998-2001. J Infect Dis. antibodies to Coccidioides immitis using a novel enzyme-linked
2005;191:1981-1987. immunosorbent assay (EIA) method. Stanford University, CA: 5th
8. Maddy KT. Disseminated coccidioidomycosis of the dog. J Am Vet International Conference on Coccidioidomycosis; 1994:137-146.
Med Assoc. 1958;132:483-489. 26. Graupmann-Kuzma A, Valentine BA, Shubitz LF, et  al. Coccidi-
9. Johnson LR, Herrgesell EJ, Davidson AP, et al. Clinical, clinicopath- oidomycosis in dogs and cats: a review. J Am Anim Hosp Assoc.
ologic, and radiographic findings in dogs with coccidioidomycosis: 2008;44:226-235.
24 cases (1995-2000). J Am Vet Med Assoc. 2003;222:461-466. 27. Greene RT. Coccidioidomycosis and paracoccidioidomycosis. In:
10. Davidson AP, Pappagianis D. Canine coccidioidomycosis: 1970 to Greene CE, ed. Infectious Diseases of the Dog and Cat. 4th ed.
1993. Stanford University, CA: 5th International Conference on St. Louis, MO: Elsevier Saunders; 2012:634-645.
Coccidioidomycosis; 1994:155-162. 28. Yturraspe DJ. Clinical evaluation of a latex particle agglutination
11. Butkiewicz CD, Shubitz LE, Dial SM. Risk factors associ- test and a gel diffusion precipitin test in the diagnosis of canine coc-
ated with Coccidioides infection in dogs. J Am Vet Med Assoc. cidioidomycosis. J Am Vet Med Assoc. 1971;158:1249-1256.
2005;226:1851-1854. 29. Kirsch EJ, Greene RT, Prahl A, et  al. Evaluation of Coccidioides
12. Greene RT, Troy GC. Coccidioidomycosis in 48 cats: a retrospec- antigen detection in dogs with coccidioidomycosis. Clin Vaccine
tive study (1984-1993). J Vet Intern Med. 1995;9:86-91. Immunol. 2012;19:343-345.
13. Beaudin S, Rich LJ, Meinkoth JH, et al. Draining skin lesion from 30. Vucicevic D, Blair JE, Binnicker MJ, et al. The utility of Coccidioi-
a desert poodle. Vet Clin Pathol. 2005;34:65-68. des polymerase chain reaction testing in the clinical setting. Myco-
14. Shubitz LE, Butkiewicz CD, Dial SM, et al. Incidence of Coccidioi- pathologia. 2010;170:345-351.
des infection among dogs residing in a region in which the organ- 31. Lauer A, Baal JD, Baal JC, et al. Detection of Coccidioides immi-
ism is endemic. J Am Vet Med Assoc. 2005;226:1846-1850. tis in Kern County, California, by multiplex PCR. Mycologia.
15. Millman TM, O’Brien TR, Suter PF, et  al. Coccidioidomyco- 2012;104:62-69.
sis in the dog: its radiographic diagnosis. J Am Vet Radiol Soc. 32. Reed RE, Hoge RS, Trautman RJ. Coccidioidomycosis in two cats.
1979;20:50-65. J Am Vet Med Assoc. 1963;143:953-956.
16. Burtch M. Granulomatous meningitis caused by Coccidioides 33. Galgiani JN, Catanzaro A, Cloud GA, et  al. Comparison of oral
immitis in a dog. J Am Vet Med Assoc. 1998;212:827-829. fluconazole and itraconazole for progressive, nonmeningeal coc-
17. Pryor Jr WH, Huizenga CG, Splitter GA, et  al. Coccidioi- cidioidomycosis. A randomized, double-blind trial. Mycoses Study
des immitis encephalitis in two dogs. J Am Vet Med Assoc. Group. Ann Intern Med. 2000;133:676-686.
1972;161:1108-1112. 34. Gautam R, Srinath I, Clavijo A, et al. Identifying areas of high risk
18. Angell JA, Merideth RE, Shively JN, et al. Ocular lesions associated of human exposure to coccidioidomycosis in Texas using serology
with coccidioidomycosis in dogs: 35 cases (1980-1985). J Am Vet data from dogs. Zoonoses Public Health, 2012.
Med Assoc. 1987;190:1319-1322. 35. Kohn GJ, Linne SR, Smith CM, et  al. Acquisition of coccidioi-
19. Shively JN, Whiteman CE. Ocular lesions in disseminated coccidi- domycosis at necropsy by inhalation of coccidioidal endospores.
oidomycosis in 2 dogs. Pathol Vet. 1970;7:1-6. Diagn Microbiol Infect Dis. 1992;15:527-530.
20. Heinritz CK, Gilson SD, Soderstrom MJ, et al. Subtotal pericardectomy 36. Stevens DA, Clemons KV, Levine HB, et al. Expert opinion: what
and epicardial excision for treatment of coccidioidomycosis-induced to do when there is Coccidioides exposure in a laboratory. Clin
effusive-constrictive pericarditis in dogs: 17 cases (1999-2003). J Am Infect Dis. 2009;49:919-923.
Vet Med Assoc. 2005;227:435-440.
21. Angell JA, Shively JN, Merideth RE, et al. Ocular coccidioidomy-
cosis in a cat. J Am Vet Med Assoc. 1985;187:167-169.
CHAPTER 64

Sporotrichosis
Jane E. Sykes

result, an S. schenckii species complex has been identified, which


Overview of Sporotrichosis includes at least six species: S. schenckii sensu stricto, Sporothrix
First Described: 1896, in Maryland, USA, by Benjamin Schenk1 brasiliensis, Sporothrix globosa, Sporothrix mexicana, Sporo-
Cause: Sporothrix schenckii complex fungi (ascomycetes, tele- thrix albicans, and Sporothrix luriei.5-7 The epidemiology of the
omorph Ophiostoma stenoceras) various members of the complex is as yet incompletely under-
stood. S. globosa has a worldwide distribution, whereas S. brasil-
Affected Hosts: Cats, dogs, humans, and a variety of other
iensis appears to be limited to Brazil. S. schenckii, S. brasiliensis,
mammalian and avian species
and S. albicans have been isolated from cats, and S. schenckii and
Geographic Distribution: Tropical and temperate zones S. luriei have been isolated from dogs.8
worldwide Cats are more susceptible to sporotrichosis than dogs and
Mode of Transmission: Cutaneous inoculation or inhalation are thought to facilitate spread of organisms in the environ-
of conidia ment.9 Contaminated claw or bite wounds, as well as autoinoc-
ulation during grooming, are thought to be important modes of
Major Clinical Signs: Cutaneous nodules or draining skin transmission.10 Male cats are overrepresented in most studies
lesions, lymphadenopathy (60% to 70% of affected cats), which may reflect the tendency
Differential Diagnoses: Other deep mycoses (especially of male cats to roam outdoors and be involved in aggressive
cryptococcosis in cats), mycobacterial infections, nocar- interactions.10-13 In Rio de Janeiro, a large outbreak of sporo-
diosis or actinomycosis, leishmaniosis, squamous cell trichosis has occurred over the past decade, which has involved
carcinoma, feline eosinophilic granuloma complex, feline more than 1000 humans, 60 dogs, and 1500 cats.1 Of affected
herpesviral dermatitis, foreign body granulomas cats in this outbreak, Sporothrix spp. were isolated from 100%
Human Health Significance: Many human infections are of 76 cutaneous lesions, 66% of 71 swab specimens from the
acquired from the environment, but direct transmission nasal cavity, 41% of 79 oral swab specimens, and 40% of 38
can follow contact between infected cats and humans, nail fragments.14 The fungus was also isolated from 3 of 84
and especially cat bites or scratches. apparently healthy cats that were in contact with affected ani-
mals. Twenty percent of cats were positive for antibodies to
FIV, 1% of cats were positive for FeLV antigen, and 1% of
cats were positive for both FIV antibodies and FeLV antigen.10
Etiology and Epidemiology More than 85% of affected cats were less than 4 years of age.
Of isolates from affected humans in the outbreak, most were
Organisms that belong to the Sporothrix schenckii species com- S. brasiliensis, but S. schenckii sensu stricto and S. globosa were
plex are dimorphic, saprophytic fungi that cause sporotrichosis, also isolated.7
a subacute to chronic disease that most often results from cuta- In dogs, sporotrichosis is relatively rare. Cutaneous intro-
neous or subcutaneous inoculation of the organism through a duction of the organism with plant foreign bodies has been sug-
puncture wound, but can also follow inhalation of the fungus. gested in some cases. In one dog, the portal of entry was thought
Sporotrichosis has been described in cats, dogs, humans, and to be an alligator bite wound.15 In the Brazilian outbreak, con-
a variety of other domestic and wild animal hosts that include tact with cats appeared to be an important mode of transmission
equine species, ruminants, swine, rats, mice, hamsters, armadil- to dogs.16 No clear breed or sex predisposition has been identi-
los, domestic fowl, chimpanzees, and dolphins. fied, and dogs in the Brazilian outbreak ranged in age from 6
Sporothrix species thrive in humid conditions (92% to 100%) months to 12 years (median, 4 years). Mixed sporotrichosis and
and at mean temperatures between 25°C and 30°C. They prefer cryptococcosis was reported in a dog from Ontario.17
moist soil that is rich in decaying vegetation, as well as sphag-
num moss, wood, thorns, or hay, where they grow as mycelia. Clinical Features
The fungus is distributed worldwide in tropical and temperate
zones but is most prevalent in tropical or subtropical regions Signs and Their Pathogenesis
of the Americas. Regions of endemicity include Mexico, Brazil, Transmission of Sporothrix primarily results from cutane-
Uruguay, Peru, Japan, India, and South Africa; sporotrichosis ous inoculation of organisms in the environment, but can also
is uncommon in Europe. Although all Sporothrix isolates have result from inhalation of conidia. Direct contact with cats that
in the past been grouped into one species, multiple genotypes have sporotrichosis (without an apparent break in the skin) can
exist that vary in virulence and geographic distribution.2-4 As a also result in transmission.10 Once within tissues, the organism

624
CHAPTER 64  Sporotrichosis 625

TABLE 64-1
Proposed Classification Scheme for Clinical Forms
of Sporotrichosis in Human Patients1
Relevance to Cats
Group Subcategory and Dogs
Cutaneous Fixed (or localized) All forms described in
Lymphocutaneous cats and dogs, but
Multifocal (or dis- fixed or ­multifocal
seminated) disease most ­common
Mucosal Ocular Occurs in cats and
Nasal dogs
Other
Extracutaneous Pulmonary Pulmonary and gen-
Osteoarticular eralized disease is
Meningeal primarily described
Generalized (or in cats. Pulmonary,
disseminated) generalized, and os-
teoarticular disease
occur rarely in dogs.
A
Special forms Spontaneous Spontaneous regression
­regression has been described
Hypersensitivity in cats and especially
(erythema nodo- dogs
sum, erythema
multiforme)

converts into a yeast form. The yeast form has a cell wall that
contains glucans, a glycoprotein fraction known as peptidor-
hamnomannan, and galactose-containing polysaccharides. The
cell wall also contains melanin, which may protect the yeast
from oxidative damage.1,9 Adhesins on the surface of the organ-
ism bind fibronectin, which appears to be important for viru-
lence.18 Organisms that are thermotolerant (able to survive at
temperatures closer to body temperature, or 37°C) may be more
likely to cause invasive disease.9
Several clinical forms of sporotrichosis are recognized in B
human patients. The clinical form of disease that develops
depends on host factors and the virulence of the infecting Spo- FIGURE 64-1  Cutaneous and mucosal sporotrichosis. A, 2-year-old female spayed
rothrix strain. A suggested classification system for human Bengal that had ulcerated lesions of the face, thoracic limb, and tail. B, Cutaneous and
sporotrichosis categorizes the distribution of lesions into cuta- conjunctival sporotrichosis in a 1-year-old male neutered domestic shorthair. (Courtesy
University of California, Davis, Veterinary Dermatology Service.)
neous, mucosal, or extracutaneous (Table 64-1). Cutaneous
lesions are crusted, plaque-like, or nodular lesions that often
ulcerate or drain serosanguineous fluid. They may be fixed inoculation. Transmission by inhalation is thought to occur in
(localized), track along the course of lymphatics (lymphocuta- some cats, because pulmonary and nasal cavity disease with
neous), or be multifocal (sometimes referred to as disseminated associated respiratory signs are common in cats.10-13,19 Respira-
cutaneous disease) (Figures 64-1 and 64-2). More than 50% tory signs include sneezing, cough, tachypnea, increased respi-
of human patients develop the lymphocutaneous form and the ratory effort, stertor, and/or nasal discharge. Respiratory signs
infection spreads proximally along lymphatics, with formation were present in 44% of 347 cats in the Brazilian outbreak10 and
of multiple secondary nodules (also known as gummata) that can also occur in dogs.13,16 Sneezing may occur before the onset
may ulcerate. This form has not been clearly recognized in cats of cutaneous lesions in cats, and some cats or dogs show only
and appears to be uncommon to rare in dogs.16 Up to 80% respiratory signs. Cats also can develop generalized disease, with
of cats with sporotrichosis have multiple, widely distributed involvement of multiple organ systems, which include the lungs,
cutaneous lesions.10,13 These may develop as a result of auto- liver, spleen, kidneys, lymph nodes, and testicles.13,20 Co-infection
inoculation during grooming, or they can be a manifestation with FIV does not appear to be a risk factor for dissemination.21
of a disseminated (or generalized) systemic infection. Isolation of Sporothrix species from the blood of 35% of 49 cats
Extracutaneous disease follows inhalation of conidia or with focal or multifocal cutaneous involvement was reported.21
hematogenous spread of Sporothrix from sites of cutaneous Clinical signs of systemic involvement include fever, generalized
626 SECTION 3  Fungal and Algal Diseases

A B
FIGURE 64-2  Multifocal ulcerated cutaneous lesions on the head (A) and left thoracic limb (B) caused by Sporothrix spp. in a 2-year-old male neutered Labrador retriever. (Courtesy
University of California, Davis, Veterinary Dermatology Service.)

TABLE 64-2
Diagnostic Assays Currently Available for Sporotrichosis in Dogs and Cats
Assay Specimen Type Target Performance
Cytologic Aspirates of cutaneous nodules, Sporothrix yeasts High sensitivity (approximately 80%) in cats. Sensitivity is
examination impression smears of skin lower in dogs.
lesions or biopsies
Fungal culture Aspirates, swabs, or biopsies of Sporothrix spp. Generally sensitive (around 75%), but false negatives can
affected tissues occur if specimen size is low. Most specific method.
Requires several weeks’ incubation. Allows species
identification via molecular methods and antifungal
drug susceptibility testing.
Histopathology Biopsy specimens from affected Sporothrix spp. Sensitivity is higher (>60%) in cats when compared with
tissues yeasts dogs (<20%). Special stains such as Gomori’s methena-
mine silver and immunohistochemical stains facilitate
organism detection and identification.

lymphadenomegaly, anorexia, dehydration, vomiting, and nodules. Lesions range in diameter from a few millimeters to
weight loss. These signs were each reported in fewer than 20% several centimeters. Extensive zones of necrosis with exposure
of cats with sporotrichosis; vomiting occurred in fewer than 5% of underlying muscle and bone have also been described.10
of the cats.10 Weight loss, vomiting, or anorexia can also occur Regional lymphadenomegaly may be detected. Other less fre-
in dogs.13,16 Other rare forms of extracutaneous sporotrichosis quent physical examination findings in cats are fever, nasal
described in humans include osteoarticular and meningeal sporo- discharge, decreased nasal airflow, tachypnea, increased respi-
trichosis. Osteoarticular sporotrichosis occurs rarely in dogs and ratory effort, dehydration, and thin body condition.
may manifest clinically as lameness and synovial effusion.12,22 In dogs, lesions consist of single or multiple ulcers or nodules
on the head, trunk, forelimbs, and/or digits (see Figure 64-2).
Physical Examination Findings Rarely, they course along lymphatics.12,13,16,24 The nasal pla-
In cats, cutaneous lesions are most often found on the head num was affected in 59% of 44 dogs in one study, and 21% of
(especially the bridge of the nose and the nasal planum, eyelids, dogs had nasal mucosal involvement.16 Regional lymphadeno-
and/or pinnae) and have a similar appearance to those caused megaly is common.
by Cryptococcus spp. (see Figure 64-1).10,12,23 Other common
sites for skin lesions are the distal limbs, digits, and tail, but Diagnosis
lesions can also be found elsewhere on the body. Swelling of
the conjunctiva or lesions in the oral cavity or nasal cavity can Diagnosis of sporotrichosis rests on identification of the organ-
be present in some affected cats (see Figure 64-1). Cutaneous ism by cytologic or histopathologic evaluation, or by fungal
or subcutaneous lesions are often firm nodules that sometimes culture of lesions (Table 64-2). A high degree of suspicion is
enlarge to form soft cutaneous masses. Nodules can ulcerate required for the disease in non-endemic regions. Sporotrichosis
and drain serosanguineous to purulent fluid, but are usually not should be considered in any dog or cat with cutaneous lesions
painful or pruritic.12 In some cases, ulcerations and draining suggestive of cryptococcosis or other deep mycoses, and espe-
skin lesions are not associated with the formation of discrete cially any cat with ulcerative or nodular lesions of the face.
CHAPTER 64  Sporotrichosis 627

FIGURE 64-4  Cytology of an impression smear from a skin lesion of the cat in Figure
64-1, A. There are large numbers of extracellular and intracellular pleomorphic yeasts, with
morphology consistent with Sporothrix spp.

Urinalysis
Urinalysis in animals with sporotrichosis usually reveals no clin-
ically significant abnormalities.

Diagnostic Imaging
Plain Radiography
Findings on plain thoracic radiographs in cats with pulmonary
sporotrichosis have not been described in detail. An interstitial
infiltrate was described in one affected cat.20 Thoracic radio-
graphs were performed on one dog and five cats seen at the
author’s teaching hospital, but no radiographic abnormalities
were present. Radiographs of bones that underlie skin lesions
typically reveal only soft tissue swelling; rarely is underlying
osteomyelitis detected.
B
FIGURE 64-3  Nasal sporotrichosis with communication to the overlying skin in a Advanced Imaging
14-year-old male neutered domestic shorthair. A, The cat had a 1-year history of sneezing and In the author’s hospital, computed tomography (CT) of the
a 2-month history of a mass on the bridge of the nose. B, Computed tomography revealed a nasal cavity in a cat with a lesion on the bridge of the nose
nasal cavity mass with nasal bone destruction and involvement of the subcutaneous tissues. revealed an irregular rim-enhancing mass within the nasal cavity
that invaded through the nasal bones to involve the subcutane-
ous tissues (Figure 64-3). Severely thickened turbinate mucosa,
an intranasal soft tissue density mass, and focal osteolysis were
Laboratory Abnormalities reported on nasal CT in a dog with signs of nasal cavity disease
Complete Blood Count due to sporotrichosis.25
In many dogs and cats with sporotrichosis, the CBC is unre-
markable. The most common abnormalities are mild anemia Microbiologic Tests
and leukocytosis due to a neutrophilia, sometimes with a mild Cytologic Examination
left shift.10,24 Monocytosis, eosinophilia, lymphocytosis, or Cytologic examination of impression smears or aspirates of skin
lymphopenia may also be present. Approximately 50% of dogs lesions typically reveals pyogranulomatous inflammation. In
in the Brazilian outbreak were anemic, and around 20% had cats, large numbers of round to cigar-shaped yeasts can be found
eosinophilia or neutrophilia.16 intracellularly within neutrophils or macrophages and extracel-
lularly (Figure 64-4). The yeasts are 4 to 6 µm in diameter and
Serum Biochemical Tests can exhibit a single bud with a narrow base. The sensitivity of
The most frequent serum biochemistry findings in cats and dogs cytologic examination of impression smears from skin lesions of
with sporotrichosis are hyperglobulinemia and hypoalbumin- 806 cats with sporotrichosis from Brazil was 79%.26 Yeasts are
emia.10,16 Rarely, increased liver enzyme activities are present usually not found in bronchoalveolar lavage fluid from cats with
in affected dogs.16 respiratory signs.19 In general, dogs have very low numbers of
628 SECTION 3  Fungal and Algal Diseases

A B C
FIGURE 64-5  Sporothrix schenckii growing on laboratory media. A, The fungus grows on fungal isolation media as a white mold at 30°C with production of pigmented conidia.
B, Using light microscopy, the mold form is characterized by the production of conidia in a flower-like arrangement. Lactophenol cotton blue stain, 1000× oil magnification. C, At 37°C,
the organism converts to the yeast form. Gram stain, 1000× oil magnification.

organisms in skin lesions, which leads to a high proportion of


false-negative results.12,16 Sporothrix spp. may be confused with
Histoplasma capsulatum, Cryptococcus spp., Candida spp.,
Leishmania infantum, and potentially other protozoan parasites
such as Toxoplasma gondii and Neospora caninum.

Fungal Culture
Definitive diagnosis of sporotrichosis can be made by isola-
tion and identification of the organism from clinical speci-
mens (such as skin biopsies, blood, nasal swabs, or respiratory
lavage specimens), preferably in both mycelial and yeast forms.
The sensitivity of isolation from skin biopsies exceeds 75% in
both dogs and cats.10,13,16 The chance of successful culture is
reduced if swab specimens of exudate instead of biopsies are
submitted. The organism grows in culture as a white mold,
which becomes brown to black as a result of production of
pigmented conidia, which are arranged in florets on a stem
(conidiophore) (Figure 64-5). Although growth can occur in FIGURE 64-6  Histopathology of a biopsy from the skin of an 11-year-old female
spayed domestic shorthair cat. Granulomatous inflammation is present with abundant
5 to 7 days, sometimes several weeks of incubation may be
intracytoplasmic fungal organisms consistent with Sporothrix spp.
required.9 When the organism is incubated on rich media such
as brain-heart infusion at 37°C, it converts to oval to cigar-
shaped yeasts. The organism can then be identified based on
light microscopic appearance of the mold and yeast forms (see Pathologic Findings
Figure 64-5, B and C). Determination of the Sporothrix species Gross pathologic findings at necropsy in cats with generalized
present requires subsequent molecular analysis by specialized sporotrichosis include multifocal, pinpoint (1 mm) white nodu-
laboratories.27 Antifungal susceptibility testing can be per- lar lesions within the lungs and lymphadenomegaly.20 Histopa-
formed, but established breakpoints are not available. Never- thology of lesions from cats usually reveals pyogranulomatous
theless, it may be of help for treatment of refractory cases (see inflammation with a variable number of neutrophils, histio-
Case Example). cytes, multinucleated giant cells, and lymphocytes, with fewer
plasma cells, eosinophils, and mast cells (Figure 64-6).12,13
Serologic Diagnosis Organized granuloma formation, with focal accumulations of
ELISA assays that detect antibody to Sporothrix cell wall anti- macrophages and sometimes an outer layer of plasma cells, is
gens have been developed for diagnosis of human and feline found in a minority (<15%) of skin biopsies from affected cats10
sporotrichosis.28,29 These assays are not widely available for and in more than 30% of skin biopsies from affected dogs.16,33
diagnosis of sporotrichosis in cats or dogs. Currently, no anti- Intracellular and extracellular yeasts are seen on histopathology
gen assays are available, but cross-reactivity with the H. capsu- in more than 60% of skin biopsies from affected cats.10,13 In
latum antigen assay occurs in human patients.30 contrast, organisms were found in biopsy specimens from fewer
than 20% of dogs with sporotrichosis.16,33 The presence of the
Molecular Diagnosis Using the Polymerase Chain Reaction Splendore-Hoeppli phenomenon (“asteroid bodies”) around
PCR assays have been described for detection of Sporothrix spp. yeasts is commonly described in human infections, but this is
in lesions.31,32 These have not been widely applied to the diag- not typically observed in dogs or cats.9 Application of stains
nosis of sporotrichosis in cats or dogs. such as periodic acid–Schiff (PAS) and especially Gomori’s
CHAPTER 64  Sporotrichosis 629

methenamine silver (GMS) increase the chance that organisms of treatments administered ranged from 1 to 5 (median, 2 treat-
are detected.12,34 However, despite the use of such stains, diag- ments per cat). Sterile abscess formation at the site of adminis-
nosis of canine sporotrichosis can be frustrating because of the tration occurred in 15% of cats.
scarcity of organisms in tissues. The use of immunohistochemis- Lastly, localized hyperthermia has been used as a safe and
try (IHC) to aid histologic diagnosis has been reported, although inexpensive method to treat fixed cutaneous and lymphocutane-
this is not readily available.34 In one study, the sensitivity of ous sporotrichosis in human patients and was used as the sole
PAS, GMS, and IHC for detection of Sporothrix spp. in dogs treatment in a cat with localized cutaneous disease.40 In that
with sporotrichosis was 20%, 44%, and 66%, respectively.34 case, a thermal bag that reached a temperature of 40°C to 42°C
The use of the stains in combination increased the sensitivity to was applied twice daily for 15 minutes. When used alone, this
over 80%. method is not likely to be effective in animals with disseminated
disease. Surgery or cryotherapy are other alternatives suggested
Treatment and Prognosis for treatment of localized sporotrichosis in human patients.35

Antimicrobial Treatment Prognosis


The treatment of choice for sporotrichosis in dogs, cats, and Treatment durations of 4 to 6 months are usually required
humans is itraconazole (see Chapter 9 for recommended doses). for successful treatment of sporotrichosis. A median time to
In human patients, use of itraconazole results in cure rates for clinical cure of 26 weeks (range, 8 to 131 weeks) was reported
cutaneous sporotrichosis that exceed 90%.35 Itraconazole resis- for 175 cats treated with itraconazole.11 Treatment should be
tance has been documented in some Sporothrix isolates from continued for a month after lesions resolve. Some cats may
animals.8 Fluconazole is considered less active than itraconazole require more than a year of antifungal drug treatment.10 In
for treatment of human sporotrichosis but was used successfully general, the prognosis for cure of sporotrichosis is good, with
to treat disseminated disease in a cat.12 Treatment of human, more than 70% of cats in some studies cured of the disease,
canine, and feline sporotrichosis with ketoconazole is more regardless of the number of skin lesions present.10,12 Lower
likely to result in relapses, and development of drug toxicity cure rates of 38% for itraconazole and 29% for ketoconazole
may limit therapy.10,12,22 In affected cats from Brazil, clinical were reported in another study.11 Co-infection with FIV does
cure was 1.3 times more likely in cats treated with itraconazole not seem to alter the likelihood of treatment success,10 but the
than in cats treated with ketoconazole.11 presence of respiratory signs was associated with a twofold
Alternative antifungal drugs for animals with refractory dis- increase in death rate.11 Relapse (or re-infection) may occur
ease include supersaturated potassium or sodium iodide, terbin- between 3 and 18 months after discontinuation of antifungal
afine, or amphotericin B. Oral administration of supersaturated drug therapy. Spontaneous regression of a persistent cutane-
potassium iodide (SSKI) for 30 days beyond complete resolu- ous lesion was also described in a cat.10 Euthanasia may result
tion of clinical signs was used to treat sporotrichosis in cats, from expense related to antifungal drug therapy or concerns
dogs, or humans before the widespread availability of itracon- for zoonotic transmission.
azole. However, treatment failures and toxic side effects that Spontaneous regression of cutaneous lesions has been
include anorexia, vomiting, diarrhea, and lethargy are common described in 15% of 33 dogs.16 Nearly 80% of dogs were
(41% of cats in one recent report).10,22,36,37 It has been recom- cured after itraconazole or ketoconazole treatment for 2 to 15
mended that the use of SSKI be reserved for fixed cutaneous, months (median, 2.5 months for itraconazole and 3.5 months
lymphocutaneous, or mucosal forms of the disease in human for ketoconazole).
patients. Because of its extremely bitter taste, potassium iodide
(KI) capsules have also been used to treat affected cats, with a Immunity and Vaccination
cure rate of 48% after 4 to 5 months.23 Inappetence occurred in
52% of cats, and a smaller percentage of cats developed weight Immunity imparted by T cells appears to be important in limit-
loss, diarrhea, or vomiting. Increased liver enzyme activities can ing the extent of sporotrichosis.9,41 There is no vaccine.
also occur. The dose range used was 2.5 to 20 mg/kg PO q24h
(median, 15 mg/kg). The dose was increased in 2.5 mg/kg incre- Prevention
ments every 5 days from an initial dose of 5 mg/kg PO q24h.
Alterations in thyroid function can be an adverse effect of KI Indoor housing and neutering may reduce sporotrichosis in cats.
treatment in humans.35 Treatment with terbinafine was success- Isolation of affected cats from other cats and dogs while they are
ful in a human with nasal sporotrichosis that failed treatment properly treated may also reduce transmission.
with KI and itraconazole.38
Although amphotericin B is effective, nephrotoxicity and Public Health Aspects
the need for parenteral administration have limited its use.
The primary indication for amphotericin B is for treatment of Many human sporotrichosis cases result from cutaneous
sporotrichosis that is refractory to itraconazole. Intralesional inoculation of organisms in soil or on plant material. Activi-
amphotericin B administration in combination with itracon- ties associated with human sporotrichosis include rose garden-
azole was used successfully to treat fixed or multifocal cutane- ing, topiary production, armadillo hunting, mining, carpentry,
ous lesions in cats that were refractory to itraconazole alone.39 forestry work, Christmas tree farming, and hay baling, and
The solution was prepared by addition of 5 mL of 2% lidocaine outbreaks have been reported after common exposure of mul-
and 5 mL of distilled water to a 50 mg vial of amphotericin B tiple individuals to a source of contaminated plant material.
(final concentration, 5 mg/mL). A total of 0.5 to 1.5 mL (median Laboratory-acquired infections have also been described.42,43
0.7 mL) was administered intralesionally once a week to once Zoonotic transmission has followed bites or scratches from
every other week, and 73% of the cats were cured. The number squirrels, horses, dogs, pigs, mules, insects, and especially cats.
630 SECTION 3  Fungal and Algal Diseases

In one study from Malaysia, slightly more human sporotrichosis 23 veterinary cases from northern California.12 Generalized
cases resulted from cat bites or scratches than from exposure extracutaneous sporotrichosis with involvement of multiple
to plant foreign bodies.44 Direct transmission can occur from organs has been described in immunocompromised humans,
infected cats to humans without an apparent break in the skin. such as those with AIDS.46,47
In the epidemic in Rio de Janeiro, disease in cats preceded that In contrast to feline sporotrichosis, canine sporotrichosis is of
in humans and dogs. Nearly 85% of affected humans reported minimal zoonotic importance. As occurs in humans, dogs have
contact with sick cats, and more than half of these individuals a paucity of organisms within tissues. There were no reports of
reported a history of a scratch or a bite. Women were more transmission to humans as a result of contact with affected dogs
likely than men to be affected, especially those involved in in the Rio de Janeiro outbreak.16
domestic activities and animal care.45 Five percent of the cases Owners of dogs and cats with sporotrichosis should be told
were veterinarians or veterinary technicians. Of cats with spo- to minimize contact with affected animals. If contact is nec-
rotrichosis from 225 residences in this outbreak, 91 people essary, gloves should be worn and proper hand washing per-
who had contact with infected cats acquired the infection.10 formed. Owners should also be informed of the potential for
In contrast, no human cases were associated with a series of exposure to organisms in the environment.6

CASE EXAMPLE Physical Examination:


Body Weight: 5.7 kg.
General: Bright, alert, responsive, and hydrated. T = 102.2°F
Signalment: “Bingo,” a 2-year-old male neutered domestic (39.0°C), HR = 140 beats/min, RR = 40 breaths/min, CRT 1 s.
longhair cat from Fairfield in northern California Integument, Ears, Eyes, Nose, and Throat: Ulcerated, firm
History: Bingo was referred to the University of California, cutaneous nodules (1 cm) were present on the left pinna and
Davis, Veterinary Medical Teaching Hospital for evaluation first digit of the left thoracic limb (0.25 cm). An ulceration
of refractory cryptococcosis. Initially Bingo’s owners noted (0.5 cm) was present on the bridge of the nose, which was
signs of snuffling, sneezing, and nasal discharge, which they markedly swollen (Figure 64-7, A). There was decreased nasal
attributed to a “cold.” One month later a lump appeared on airflow bilaterally that was more severe on the left side, and
Bingo’s nose. Bingo was taken to a local veterinary clinic mild left-sided mucoid nasal discharge. Marked soft tissue
where cytologic examination of an impression smear of swelling occluded the left nares. There was moderate, left-
the mass revealed organisms that were suspected to be sided serous ocular discharge. No lesions were detected on
Cryptococcus spp. A CBC showed no abnormalities. The fundoscopic examination.
owners declined treatment with itraconazole because Musculoskeletal: Body condition score was 6/9, ambulatory.
of financial limitations, so fluconazole was prescribed Cardiovascular: No clinically significant abnormalities were
(10 mg/kg PO q12h). Despite this treatment, the respiratory detected.
signs persisted, nodular skin lesions appeared on the face, Respiratory: There was a mild increase in respiratory effort
and mandibular lymphadenopathy developed. Bingo had with stertor. Thoracic auscultation revealed referred upper
been an indoor-outdoor cat but had been housed exclusively airway sounds.
indoors for the previous few months. He lived with five other Gastrointestinal and Genitourinary: Abdominal palpation
indoor cats and a dog, all of which were well. His appetite and was unremarkable. A small amount of dried feces and a
activity level had been normal and he was fed a commercial tapeworm proglottid protruded from the anus.
dry cat food. According to the owners, Bingo had tested Lymph Nodes: Only the left mandibular lymph node was
negative for FeLV and FIV approximately 1 year previously. enlarged (2 cm in diameter).

A B C
FIGURE 64-7  Effect of treatment with itraconazole and amphotericin B in a 2-year-old male neutered domestic longhair cat with multifocal cutaneous and mucosal sporotrichosis.
A, Before institution of treatment. B, After itraconazole and eight treatments with amphotericin B. C, After an additional 7 months of itraconazole treatment.
CHAPTER 64  Sporotrichosis 631

Microbiologic Testing: Latex agglutination for Cryptococcus eight amphotericin B treatments, the lesions had halved
antigen: Negative. in size (Figure 64-7, B). An additional three treatments
Cytologic examination of impression smears from the nares: were administered, after which a mild increase in BUN
Erythrocytes, neutrophils, and occasional epithelial cells concentration developed (43 mg/dL, reference range 18-33
were present. No fungal elements were identified. mg/dL). Serum creatinine was within the reference range
Histopathology of a biopsy from the nasal cavity: Pyogranulo- (1.4 mg/dL, reference range 1.1-2.2 mg/dL). The serum
matous inflammation with intralesional fungi with morphol- itraconazole concentration was 4.03 µg/mL. A new lesion
ogy consistent with Sporothrix spp. appeared on the right distal thoracic limb, and impression
Diagnosis: Sporotrichosis with nasal and multifocal cutaneous smears of the lesion revealed large numbers of Sporothrix
involvement. yeasts. The itraconazole dose was increased to 5 mg/kg PO
Treatment: Bingo was treated with a compounded form of q12h, but was reduced by the owner to 5 mg/kg PO q48h after
itraconazole (5 mg/kg PO q12h). Clinical improvement did 5 months because Bingo’s appetite was slightly decreased.
not occur after 2 weeks, so terbinafine was added (30 mg/ When the cat was reevaluated 7 months after completion of
kg PO q24h). One month later, two nodules appeared on amphotericin treatment, lesions had improved dramatically
the conjunctiva of the left eye, and the lesions on the left (Figure 64-7, C). The owner was encouraged to increase the
pinna, left thoracic limb, and nasal bridge had enlarged. Mild dose of itraconazole to 5 mg/kg PO q24h, but the cat was
subcutaneous emphysema of the face was also apparent subsequently lost to follow-up.
during respiration. The cat was bright and appetent, but Comments: This cat had nasal and multifocal cutaneous
body weight had decreased to 5.1 kg. No abnormalities sporotrichosis that was somewhat refractory to treatment
were detected on the CBC and serum biochemistry panel. with antifungal drugs. The disease was initially mistaken
Large numbers of Sporothrix yeasts were seen on impression for cryptococcosis. Limited client financial resources and
smears of the thoracic limb lesion. Fungal culture yielded compliance may have influenced treatment outcome in this
S. schenckii sensu stricto (identified at the University of case; costs of fungal culture, identification, and antifungal
Texas Health Science Center at San Antonio based on ITS, susceptibility testing were not charged to the client.
D1/D2, and calmodulin gene sequencing). Antifungal Fluconazole was ineffective, and the antifungal susceptibility
drug susceptibility testing revealed minimum inhibitory panel suggested resistance to this azole. The use of
concentrations of 1, 2, 0.06, greater than 64, 1, 1, and 16 µg/ compounded itraconazole resulted in suboptimal serum drug
mL for amphotericin B, 5-flucytosine, terbinafine, fluconazole, concentrations. However, once treatment with amphotericin B,
itraconazole, posaconazole, and voriconazole, respectively. A a brand name itraconazole formulation, and heat compression
serum itraconazole concentration was less than 0.31 µg/mL. of lesions was instituted, gradual clinical improvement
Subsequently the cat was hospitalized and treated with occurred. The change in itraconazole formulation alone might
itraconazole (Sporanox) solution (3 mg/kg PO q12h) and have been sufficient to cause disease resolution. The owners
deoxycholate amphotericin B (0.25 mg/kg IV). Terbinafine were told to wear gloves when handling the cat and to wash
treatment was discontinued. Cutaneous lesions were also their hands afterward. No disease was reported in the owners
treated with local hyperthermia (15 minutes q8h). After or any other animals in the household.

SUGGESTED READINGS 5. Romeo O, Scordino F, Criseo G. New insight into molecular phy-
logeny and epidemiology of Sporothrix schenckii species complex
Barros MB, de Almeida Paes R, Schubach AO. Sporothrix schenckii and based on calmodulin-encoding gene analysis of Italian isolates.
sporotrichosis. Clin Microbiol Rev. 2011;24:633-654. Mycopathologia. 2011;172:179-186.
Crothers SL, White SD, Ihrke PJ, et al. Sporotrichosis: a retrospective 6. Marimon R, Serena C, Gene J, et al. In vitro antifungal susceptibili-
evaluation of 23 cases seen in northern California (1987-2007). Vet ties of five species of Sporothrix. Antimicrob Agents Chemother.
Dermatol. 2009;20:249-259. 2008;52:732-734.
Vasquez-del-Mercado E, Arenas R, Padilla-Desgarenes C. Sporotricho- 7. Oliveira MM, Almeida-Paes R, Muniz MM, et  al. Pheno-
sis. Clin Dermatol. 2012;30:437-443. typic and molecular identification of Sporothrix isolates from
an epidemic area of sporotrichosis in Brazil. Mycopathologia.
REFERENCES 2011;172:257-267.
1. Lopes-Bezerra LM, Schubach A, Costa RO. Sporothrix schenckii 8. Oliveira DC, Lopes PG, Spader TB, et al. Antifungal susceptibilities
and sporotrichosis. An Acad Bras Cienc. 2006;78:293-308. of Sporothrix albicans, S. brasiliensis, and S. luriei of the S. schenckii
2. Brito MM, Conceicao-Silva F, Morgado FN, et al. Comparison of complex identified in Brazil. J Clin Microbiol. 2011;49:3047-3049.
virulence of different Sporothrix schenckii clinical isolates using 9. Barros MB, de Almeida Paes R, Schubach AO. Sporothrix schenckii
experimental murine model. Med Mycol. 2007;45:721-729. and sporotrichosis. Clin Microbiol Rev. 2011;24:633-654.
3. Kong X, Xiao T, Lin J, et al. Relationships among genotypes, vir- 10. Schubach TM, Schubach A, Okamoto T, et  al. Evaluation of an
ulence and clinical forms of Sporothrix schenckii infection. Clin epidemic of sporotrichosis in cats: 347 cases (1998-2001). J Am
Microbiol Infect. 2006;12:1077-1081. Vet Med Assoc. 2004;224:1623-1629.
4. Zhang Z, Liu X, Lv X, et  al. Variation in genotype and higher 11. Pereira SA, Passos SR, Silva JN, et  al. Response to azolic anti-
virulence of a strain of Sporothrix schenckii causing disseminated fungal agents for treating feline sporotrichosis. Vet Rec.
cutaneous sporotrichosis. Mycopathologia. 2011;172:439-446. 2010;166:290-294.
632 SECTION 3  Fungal and Algal Diseases

12. Crothers SL, White SD, Ihrke PJ, et al. Sporotrichosis: a retrospec- 29. Bernardes-Engemann AR, Costa RC, Miguens BR, et  al. Devel-
tive evaluation of 23 cases seen in northern California (1987-2007). opment of an enzyme-linked immunosorbent assay for the sero-
Vet Dermatol. 2009;20:249-259. diagnosis of several clinical forms of sporotrichosis. Med Mycol.
13. Madrid IM, Mattei AS, Fernandes CG, et al. Epidemiological find- 2005;43:487-493.
ings and laboratory evaluation of sporotrichosis: a description of 30. Assi M, Lakkis IE, Wheat LJ. Cross-reactivity in the Histoplasma
103 cases in cats and dogs in southern Brazil. Mycopathologia. antigen enzyme immunoassay caused by sporotrichosis. Clin Vac-
2012;173:265-273. cine Immunol. 2011;18:1781-1782.
14. Schubach TM, de Oliveira Schubach A, dos Reis RS, et  al. Spo- 31. Hu S, Chung WH, Hung SI, et al. Detection of Sporothrix schenckii
rothrix schenckii isolated from domestic cats with and with- in clinical samples by a nested PCR assay. J Clin Microbiol.
out sporotrichosis in Rio de Janeiro, Brazil. Mycopathologia. 2003;41:1414-1418.
2002;153:83-86. 32. Kano R, Watanabe K, Murakami M, et al. Molecular diagnosis of
15. Moriello KA, Franks P, Delany-Lewis D, et  al. Cutaneous-lym- feline sporotrichosis. Vet Rec. 2005;156:484-485.
phatic and nasal sporotrichosis in a dog. J Am Anim Hosp Assoc. 33. de Miranda LH, Quintella LP, dos Santos IB, et al. Histopathology
1987;24:621-626. of canine sporotrichosis: a morphological study of 86 cases from
16. Schubach TM, Schubach A, Okamoto T, et  al. Canine sporotri- Rio de Janeiro (2001-2007). Mycopathologia. 2009;168:79-87.
chosis in Rio de Janeiro, Brazil: clinical presentation, laboratory 34. Miranda LH, Quintella LP, Menezes RC, et  al. Evaluation of
diagnosis and therapeutic response in 44 cases (1998-2003). Med immunohistochemistry for the diagnosis of sporotrichosis in dogs.
Mycol. 2006;44:87-92. Vet J. 2011;190:408-411.
17. Shany M. A mixed fungal infection in a dog: sporotrichosis and 35. Vasquez-del-Mercado E, Arenas R, Padilla-Desgarenes C. Sporotri-
cryptococcosis. Can Vet J. 2000;41:799-800. chosis. Clin Dermatol. 2012;30:437-443.
18. Teixeira PA, de Castro RA, Nascimento RC, et  al. Cell surface 36. Burke MJ, Grauer GF, Macy DW. Successful treatment of cutaneo-
expression of adhesins for fibronectin correlates with virulence in lymphatic sporotrichosis in a cat with ketoconazole and sodium
Sporothrix schenckii. Microbiology. 2009;155:3730-3738. iodide. J Am Anim Hosp Assoc. 1983;19:542-547.
19. Leme LR, Schubach TM, Santos IB, et al. Mycological evaluation 37. Anderson NV, Ivoghli D, Moore WS, et al. Cutaneous sporotricho-
of bronchoalveolar lavage in cats with respiratory signs from Rio sis in a cat: a case report. J Am Anim Hosp Assoc. 1973;9:526-529.
de Janeiro, Brazil. Mycoses. 2007;50:210-214. 38. Heidrich D, Stopiglia CD, Senter L, et  al. Successful treatment
20. Schubach TM, Schubach Ade O, Cuzzi-Maya T, et  al. Pathol- of terbinafine in a case of sporotrichosis. An Bras Dermatol.
ogy of sporotrichosis in 10 cats in Rio de Janeiro. Vet Rec. 2011;86:S182-S185.
2003;152:172-175. 39. Gremião I, Schubach T, Pereira S, et  al. Treatment of refractory
21. Schubach TM, Schubach A, Okamoto T, et  al. Haematogenous feline sporotrichosis with a combination of intralesional ampho-
spread of Sporothrix schenckii in cats with naturally acquired spo- tericin B and oral itraconazole. Aust Vet J. 2011;89:346-351.
rotrichosis. J Small Anim Pract. 2003;44:395-398. 40. Honse CO, Rodrigues AM, Gremiao ID, et al. Use of local hyper-
22. Goad DL, Goad ME. Osteoarticular sporotrichosis in a dog. J Am thermia to treat sporotrichosis in a cat. Vet Rec. 2010;166:208-209.
Vet Med Assoc. 1986;189:1326-1328. 41. Carlos IZ, Sassa MF, da Graca Sgarbi DB, et al. Current research
23. Reis EG, Gremiao ID, Kitada AA, et  al. Potassium iodide cap- on the immune response to experimental sporotrichosis. Myco-
sule treatment of feline sporotrichosis. J Feline Med Surg. pathologia. 2009;168:1-10.
2012;14:399-404. 42. Cooper CR, Dixon DM, Salkin IF. Laboratory-acquired sporotri-
24. Sykes JE, Torres SM, Armstrong PJ, et  al. Itraconazole for treat- chosis. J Med Vet Mycol. 1992;30:169-171.
ment of sporotrichosis in a dog residing on a Christmas tree farm. 43. Thompson DW, Kaplan W. Laboratory-acquired sporotrichosis.
J Am Vet Med Assoc. 2001;218:1440-1443:1421. Sabouraudia. 1977;15:167-170.
25. Whittemore JC, Webb CB. Successful treatment of nasal sporotri- 44. Tang MM, Tang JJ, Gill P, et al. Cutaneous sporotrichosis: a six-
chosis in a dog. Can Vet J. 2007;48:411-414. year review of 19 cases in a tertiary referral center in Malaysia. Int
26. Pereira SA, Menezes RC, Gremiao ID, et  al. Sensitivity of cyto- J Dermatol. 2012;51:702-708.
pathological examination in the diagnosis of feline sporotrichosis. 45. Barros MB, Schubach AO, Schubach TM, et  al. An epidemic of
J Feline Med Surg. 2011;13:220-223. sporotrichosis in Rio de Janeiro, Brazil: epidemiological aspects of
27. de Oliveira MM, Sampaio P, Almeida-Paes R, et al. Rapid identi- a series of cases. Epidemiol Infect. 2008;136:1192-1196.
fication of Sporothrix species by T3B fingerprinting. J Clin Micro- 46. Freitas DF, de Siqueira Hoagland B, do Valle AC, et al. Sporotri-
biol. 2012;50:2159-2162. chosis in HIV-infected patients: report of 21 cases of endemic spo-
28. Fernandes GF, Lopes-Bezerra LM, Bernardes-Engemann AR, et al. rotrichosis in Rio de Janeiro, Brazil. Med Mycol. 2012;50:170-178.
Serodiagnosis of sporotrichosis infection in cats by enzyme-linked 47. Hardman S, Stephenson I, Jenkins DR, et al. Disseminated Sporo-
immunosorbent assay using a specific antigen, SsCBF, and crude thrix schenckii in a patient with AIDS. J Infect. 2005;51:e73-e77.
exoantigens. Vet Microbiol. 2011;147:445-449.
CHAPTER 65

Aspergillosis
Jane E. Sykes

accurate identification of Aspergillus species (especially rare spe-


Overview of Aspergillosis cies) requires advanced molecular techniques, organisms can be
First Described: Invasive aspergillosis was first described in misidentified based on their morphology in culture alone. As a
1815 in a jackdaw bird (Mayer)1 result, use of the term “species complex” is recommended when
Cause: Aspergillus spp. (an ascomycete, teleomorphs Neosar- molecular techniques are not used for identification purposes.15
torya, Emericella, Eurotium) Because the etiology, epidemiology, clinical signs, diagnosis,
and treatment of upper respiratory tract (URT) aspergillosis,
Affected Hosts: Dogs, cats, humans, and a variety of other
systemic aspergillosis, and keratomycosis/otomycosis differ,
animal species
they are considered separately in this chapter.
Geographic Distribution: Worldwide
Mode of Transmission: Inhalation of conidia from the Upper Respiratory Tract Aspergillosis
environment
Major Clinical Signs: Sinonasal aspergillosis: nasal discharge,
Etiology and Epidemiology
sneezing, epistaxis, depigmentation of the nares in dogs.
In dogs, SNA is a subacute to chronic, noninvasive disease
Sino-orbital aspergillosis (especially cats): exophthalmos,
of the nasal cavity and sinuses. It is most often caused by
mass lesions or ulcers in the pterygopalatine fossa. Dis-
­Aspergillus fumigatus. Rarely, infection by other species, such
seminated aspergillosis: inappetence, weight loss, spinal
as A­ spergillus niger complex organisms (such as Aspergillus
pain, lameness, pelvic limb paresis or paralysis, cough,
tubingensis) or Aspergillus flavus and the closely related fun-
vomiting.
gus Penicillium, occurs. SNA occurs worldwide. Any breed of
Differential Diagnoses: Sinonasal aspergillosis: neoplasia, dog can be affected, but disease is most common in large, non-
inflammatory rhinitis, oronasal fistulas, cryptococcosis, brachycephalic breeds, especially German shepherd dogs, Rott-
nasal mites (dogs), foreign bodies, and polyps (cats). Dis- weilers, Border collies, Rhodesian ridgebacks, and retrievers.
seminated aspergillosis: bacterial discospondylitis, bacte- Dogs of any age can be affected (median age around 6 years).16
rial pyelonephritis, and other systemic fungal infections, No clear sex predisposition exists. SNA is thought to occur
especially paecilomycosis and penicilliosis. when local immune mechanisms fail as a result of uncharac-
Human Health Significance: Human Aspergillus infections are terized genetic or acquired immunodeficiency syndromes, and
acquired from the environment. Direct transmission from occasionally as a result of implantation of a foreign body or
affected dogs and cats to humans does not occur. other preexisting nasal disease, such as nasal neoplasia. Rarely,
it occurs in conjunction with other systemic diseases such as
hyperadrenocorticism, diabetes mellitus, or systemic autoim-
mune disorders. However, in most cases, affected dogs are

A
spergillosis is the disease caused by pathogenic fungi that otherwise systemically well and have no evidence of immune
belong to the genus Aspergillus. Aspergillus species are compromise. Impaired cell-mediated immune responses have
ubiquitous, saprophytic, hyaline molds, the spores of been detected in dogs with SNA, but it is unclear whether this is
which can be found in soil, water, air, and decaying vegetation. a cause or consequence of Aspergillus infection.17
It is estimated that an individual person inhales several hundred Upper respiratory tract aspergillosis is relatively uncommon in
Aspergillus spores each day.2 Aspergillus species cause a variety cats, and includes SNA and SOA. Approximately 50 cases of URT
of noninvasive, semi-invasive, and invasive forms of disease in aspergillosis have been reported in cats to date.3 Fungi that belong
dogs and cats that include keratomycosis, fungal otitis externa, to the A. fumigatus complex and, rarely, other Aspergillus species
sinonasal aspergillosis (SNA), sino-orbital aspergillosis (SOA), may also be involved (see Table 65-1). A novel organism with a
bronchopulmonary aspergillosis, and disseminated aspergillosis. proposed name of Aspergillus felis sp. nov. appears to cause SOA
Aspergillus species that can cause disease in dogs and cats are in cats,12 whereas SNA is caused predominantly by A. fumigatus
shown in Table 65-1.3-14 Teleomorphs (sexual forms) of the fungus sensu stricto.3 Brachycephalic breeds such as Persians and Hima-
have been identified for some Aspergillus species. Currently, these layan Persians appear to be predisposed to SNA and especially
are named differently and include fungi that belong to the genera to SOA, but both conditions occur in any breed of cat. Affected
Neosartorya, Emericella, and Eurotium. The name Aspergillus cats range in age from 1 to 13 years (median, 5 years). No clear
may be retained even when teleomorphs are isolated from lesions, sex predisposition exists.3 Most cats with URT aspergillosis test
to avoid confusion through the use of unfamiliar names.12 Because negative for retrovirus infection; only one cat with SNA in the

633
634 SECTION 3  Fungal and Algal Diseases

TABLE 65-1
Aspergillus Species Associated with Disease in Dogs and Cats3-14
Clinical Form Host Species Aspergillus species*
Keratomycosis Cats A. fumigatus
A. flavus
Dogs Not reported
Otomycosis Cats A. fumigatus
Dogs (very rare) A. ochraceus
A. versicolor
A. niger
Sinonasal Dogs A. fumigatus
aspergillosis A. niger
A. nidulans
A. flavus
A. tubingensis
Cats A. fumigatus FIGURE 65-1  Unilateral depigmentation and ulceration of the nares of a 7-year-old
shepherd mix with sinonasal aspergillosis. (Courtesy Dr. Lynelle Johnson, Small Animal
A. niger
Internal Medicine Service, University of California, Davis.)
A. udagawae
A. flavus
Neosartorya spp. ciliostasis and apoptosis and can suppress the host immune
Sino-orbital Cats A. felis sp. nov. response.2 The fungus also produces proteases and phospholi-
aspergillosis pases. Chemokines such as Il-8 and monocyte chemoattractant
protein-1 (MCP-1/CCL2) are secreted by host inflammatory cells
Bronchopulmonary Dogs and cats A. fumigatus in response to the fungus, which may recruit neutrophils and mac-
aspergillosis A. flavus (dog) rophages to the site of infection.20 Severe disease is associated with
Disseminated Dogs A. terreus destruction of nasal bones, the cribriform plate, and/or the orbit.
aspergillosis A. deflectus Clinical signs of SNA in dogs and cats consist of sneezing,
A. fumigatus reverse sneezing, serous to mucopurulent nasal discharge, depig-
A. niger mentation of the nasal planum, and, less commonly, epistaxis.
A. flavipes Nasal discharge is initially unilateral but can become bilateral as
A. lentulus disease progresses. Animals with advanced disease can be lethar-
A. carneus gic and inappetent and may lose weight. Nasal pain can mani-
A. versicolor fest in dogs as pawing at the face, hiding, or withdrawal when
A. alabamensis approached. Destruction of the nasolacrimal duct or invasion of
A. felis sp. nov. the orbit can lead to epiphora. Osteolysis of the cribriform plate
Cats A. fumigatus is often subclinical, but occasionally leads to meningitis with
neurologic signs such as obtundation or seizures.
*Some isolates have not been identified beyond the species complex In cats with SOA, Aspergillus invades the submucosal tissue
level (including some A. flavus, A. fumigatus, and A. niger). of the nasal cavity and sinus and extends through the orbital
bone and into the retrobulbar space. A profound granulomatous
inflammatory response to the fungus results in the formation of
literature was co-infected with FeLV.18 Five of 8 cats with SNA a retrobulbar mass, with clinical signs that include exophthal-
evaluated at the author’s teaching hospital had diabetes mellitus.4 mos, protrusion of the third eyelid, mass lesions in or ulceration
of the pterygopalatine fossa, and, rarely, blindness due to inva-
Clinical Features sion of the optic nerve or optic chiasm.3,21-23 Inappetence and
dysphagia can also develop.22 Occasionally, the clinical signs
Signs and Their Pathogenesis of SOA develop after initial signs of SNA, such as sneezing and
Aspergillus conidia (or spores), which are 2 to 3 µm in diameter, nasal discharge.3,21
are inhaled and deposit in the nasal cavity and/or sinuses, where
they adhere to epithelial cells. In the absence of adequate immune Physical Examination Findings
defenses, the conidia enlarge and germinate to form hyphae. SNA Physical examination findings in dogs with SNA include unilat-
is a noninvasive disease, and the fungus does not extend beyond eral or bilateral serous or mucopurulent nasal discharge, epistaxis,
the mucosal epithelium.19 Production of toxins by the fungus and pain on palpation of the face, mild mandibular lymphadenomeg-
a profound host inflammatory response is thought to explain aly, and sometimes depigmentation and ulceration of the nares
the extensive turbinate and bony destruction that follows fungal (Figure 65-1).24,25 Nasal airflow is usually normal or increased
colonization. Toxins produced by A. fumigatus include aflatoxin, because of turbinate lysis. Serous ocular discharge occurs uncom-
hemolysin, fumagillin, and gliotoxin,2 but the role that these tox- monly as a result of destruction of the bones of the orbit. Rarely,
ins play in canine SNA has not been determined. Gliotoxin induces palpation of the nasal bones or the hard palate can reveal defects
CHAPTER 65  Aspergillosis 635

A B

C D
FIGURE 65-2  A, Seven-year-old male neutered Scottish fold cat with exophthalmos, conjunctivitis, and blindness of the right eye secondary to sino-orbital aspergillosis. A 1- to 2-cm
mass was also identified in the pterygopalatine fossa on oral examination. B-D, MRI image of the head of the same cat. The right eye was displaced rostrally by a large retrobulbar mass that
is mildly, heterogeneously hyperintense on T1-weighted images (B), hyperintense on T2-weighted images (C), and enhanced with contrast (D). The mass extends medial to the ramus of
the mandible and enters the nasopharynx as well as the oropharynx. A small T1-hyperintense structure is noted in the left frontal sinus.

in the underlying bone. Dogs with cribriform plate involvement Diagnosis


and meningitis may be obtunded or dehydrated. Dogs with severe
epistaxis can have evidence of melena on rectal examination sec- Early diagnosis of URT aspergillosis relies on a high degree
ondary to swallowed blood. Most affected dogs are afebrile. of suspicion for the disease and availability of client finan-
Physical examination findings in cats with SNA consist of cial resources, because there is a lack of sensitive, noninvasive
serous or mucopurulent nasal discharge, sneezing, stertor, epi- diagnostic tests for the disease. Definitive diagnosis requires
staxis, and, in some cases, facial distortion.3 Cats with SOA a combination of imaging, visualization of fungal plaques by
usually have unilateral exophthalmos and deviation of the rhinoscopy/sinuscopy, and identification of fungal structures
globe; bilateral involvement has also been described (Figure on cytology or histopathology. Imaging should be performed
65-2).3,22,23 There may be resistance to ocular retropulsion and before rhinoscopy to avoid the influence of iatrogenic hemor-
protrusion of the third eyelid. Miosis, an absent pupillary light rhage on the results.
response, and an absent menace response may be present. On
examination of the oral cavity, most affected cats also have Laboratory Abnormalities
a mass or ulcer in the pterygopalatine fossa ipsilateral to the Dogs with SNA often have no abnormalities on the CBC, serum
affected eye. Other less common findings are exposure keratitis chemistry profile, or urinalysis. Occasionally, mild nonregen-
and corneal ulceration, mandibular lymphadenomegaly, pain erative anemia; mild neutrophilia, rarely with a left shift; mild
on opening the mouth, and/or ulceration of the hard palate.3,22 eosinophilia; and/or mild hypoalbuminemia are present.4 Cats
636 SECTION 3  Fungal and Algal Diseases

A B

C D
FIGURE 65-3  Plain radiographs (A and B) and CT scan images (C and D) of the nasal cavity of a 6-year-old female spayed golden retriever with sinonasal aspergillosis. A, Open-mouth
view. There is marked loss of detail in the left nasal cavity. B, Skyline projection of the frontal sinuses. There is increased opacity in the left frontal sinus. C, A transverse image through the
nasal cavity shows marked turbinate loss on the left side. D, Transverse image through the frontal sinuses and region of the cribriform plate. There is moderate fluid accumulation and irregu-
lar soft tissue structures in the left frontal sinus, and the frontal bone that overlies the sinus is thickened and irregular. Bony destruction at the level of the cribriform plate is present (arrow).

with SOA may have a mild mature neutrophilia and sometimes best appreciated on CT. Findings on CT or MRI in canine and
mild to severe hyperglobulinemia.3,21 feline SNA include loss of nasal turbinates and defects in the
nasal septum; increased soft tissue opacity within the nasal cavity
Diagnostic Imaging and/or sinuses due to mucosal inflammation, fluid accumulation,
Advanced Imaging or fungal plaques; hyperostosis of the frontal bones; and man-
Whenever possible, computed tomography (CT) or MRI are pre- dibular or retropharyngeal lymph node enlargement.3,26-29 Oste-
ferred imaging modalities for the nasal cavity because of their olysis of the cribriform plate, orbit, frontal bones, and, rarely,
increased sensitivity over plain radiography for evaluation of the the maxillary or palatine bones may be present (Figure 65-3, C).
nasal cavity, sinuses, and cribriform plate.26 Bony changes are Meningeal enhancement is sometimes evident on contrast
CHAPTER 65  Aspergillosis 637

Microbiologic Tests
Cytologic Examination
Cytologic examination of blind or rhinoscopy-directed mucosal
swabs, brush specimens of the nasal cavity, or nasal biopsies
from dogs or cats with SNA can reveal Aspergillus hyphae and
sometimes also conidia. Hyphae stain well with Wright’s stains.
They are septate and branch at 45-degree angles (see Figure
4-3). Large numbers of degenerate neutrophils, ciliated epithe-
lial cells, and/or bacteria are also usually present. In one study,
the sensitivity of cytologic examination was 93% when cytol-
ogy brush specimens were examined and 100% when squash
preparations of biopsies were examined, but it was 20% or less
when nasal discharge or smears made from blind swab speci-
mens were examined.31
Cytologic examination of ultrasound- or CT-guided aspi-
rates of retrobulbar masses from cats with SOA reveals a mixed
inflammatory response and sometimes fungal hyphae.

Fungal Culture
Aspergillus fumigatus can be readily grown on routine labora-
tory media. Colonies of mold generally appear within 96 hours,
but occasionally longer incubation times are required.32 In the
FIGURE 65-4  Rhinoscopic view of a large gray fungal plaque in the nasal cavity of an absence of supportive rhinoscopic, cytologic, or histopathologic
8-year-old female pit bull terrier with sinonasal aspergillosis. findings, positive cultures from the nasal cavity (or other non-
sterile sites) require cautious interpretation because Aspergillus
administration in dogs with cribriform plate destruction.25 Fun- may be found within the URT of healthy animals. Nevertheless,
gal plaques and mucosal secretions fail to enhance with contrast in two separate studies, no dogs with nonfungal nasal disor-
and cannot be readily distinguished from each other. In some ders had positive culture results for A ­ spergillus.16,32 In one of
dogs, lesions are limited to the frontal sinus.27 these studies (which included 21 dogs with SNA and 37 dogs
Advanced imaging of the head in cats with SOA reveals a with nonfungal nasal disease), the sensitivity of fungal culture
retrobulbar mass that enhances diffusely or heterogeneously of nasal biopsies for diagnosis of canine SNA was 81%.16 In
with contrast. Displacement of the globe, osteolysis of the orbit, the other study, fungal culture of biopsy specimens or plaque
mandibular lymphadenomegaly, and changes consistent with specimens collected by rhinoscopic guidance (≥75%) was more
SNA can also be present. With MRI, retrobulbar masses are sensitive than culture of blindly collected nasal swab specimens
T2-hyperintense. Invasion of temporal and masseter muscles by (<20%).32
the mass may also be apparent on MRI (see Figure 65-2).30 Growth of an A. fumigatus complex organism from guided
aspirates or biopsy specimens collected from a retrobulbar mass
Plain Radiography in a cat strongly suggests a diagnosis of SOA. In one study, fun-
Plain skull radiographs in dogs with SNA may show unilateral gal culture of nasal biopsies or aspirates from cats with SOA
or bilateral loss of turbinate detail, destruction of the nasal sep- and SNA had a sensitivity greater than 95%.3
tum, and/or increased soft tissue opacity within the nasal cav-
ity. The open-mouth view provides optimum evaluation of the Serologic Diagnosis
turbinates (see Figure 65-3, A). A “skyline” view of the nasal Both serum antibody and antigen assays have been evaluated
sinuses may show opacification of one or both sinuses and fron- for diagnosis of URT aspergillosis. Assays for detection of
tal bone hyperostosis (see Figure 65-3, B).26 serum antibodies to Aspergillus include ELISA and gel immuno-
diffusion (ID) assays (see Figure 2-7). Assay types vary in their
Rhinoscopy and Sinuscopy sensitivity and specificity for diagnosis of SNA, and clinical per-
Visualization of fungal plaques by rhinoscopy and sinuscopy con- formance data for some assays used in commercial veterinary
firms a diagnosis of SNA. Plaques are white, gray, yellow, black, diagnostic laboratories may not be available. The sensitivity of
greenish, or a mixture of these colors and are adhered to the nasal one gel ID assay (Meridian Bioscience) for diagnosis of canine
mucosa or mucosa of the frontal sinus (Figure 65-4). Caudal rhi- SNA was 57% to 77%,16,25,33 and the specificity was at least
noscopy is performed with a flexible endoscope, after which a 98%.16,33 The sensitivity and specificity of another gel ID assay
rigid telescope or flexible endoscope is used to examine the ros- (Immuno-Mycologics, OK) was 31% and 97%, respectively.34
tral portions of the nasal cavity. In some cases, extensive turbi- The sensitivity and specificity of an ELISA that detects antibod-
nate destruction allows a flexible endoscope to be passed directly ies against a purified Aspergillus antigen extract was 88% and
into the frontal sinuses through the nasofrontal opening. If this 97%, respectively.33 Thus, negative results with these antibody
is not possible, but CT or MRI findings suggest the possibility of assays do not rule out SNA, but positive results strongly suggest
frontal sinus lesions, trephination or surgical exploration of the the disease is present. The sensitivity and specificity of serum
frontal sinuses is necessary to visualize (and treat) this region. antibody assays for diagnosis of feline URT aspergillosis has not
After trephination, a rigid endoscope can be used to examine the been evaluated in a large number of cats, but both positive and
frontal sinus through the trephination site. negative assay results have been reported in affected cats.3,28,35
638 SECTION 3  Fungal and Algal Diseases

A B
FIGURE 65-5  Histopathology of a nasal biopsy from a 4-year-old intact male Border collie with sinonasal aspergillosis. A, There is a profound neutrophilic inflammatory response
beneath a thick mat of fungal hyphae, which stain magenta. Periodic acid–Schiff stain, 400× magnification. B, Aspergillus hyphae (arrowheads), conidiophores (small arrows) and conidia
(spores) can be seen on the surface of the lesion. H&E stain, 1000× oil magnification.

A serum Aspergillus galactomannan antigen ELISA assay (epithelioid macrophages, eosinophils, neutrophils, lymphocytes,
(Platelia), originally developed for diagnosis of invasive asper- and plasma cells) and a peripheral zone of fibrosis surround the
gillosis in humans, has been studied for the diagnosis of SNA granulomas. Inflammatory lesions may efface muscle and bone.
and SOA in dogs and cats, but its sensitivity and specificity have
been low.5,33,36,37 Only 4 of 17 dogs with SNA were positive in Treatment and Prognosis
one study, and only weak positive results were obtained (galac-
tomannan indices 0.5-0.8, reference range <0.5).33 False positive Successful treatment of SNA is challenging. Because of the large size
results occurred in 11 of 62 dogs without SNA (galactomannan of the nasal cavity and sinuses, clinical signs are often not apparent
indices of 0.5-1.5). Only 3 of 13 cats with URT aspergillosis until disease is advanced. Delays in diagnosis can occur because
tested positive, and false-positive results were obtained in a of the costly and invasive nature of diagnosis and treatment and
number of cats without URT aspergillosis (specificity 78%).38 the need for specialized imaging techniques and endoscopic exper-
tise. Owners of adult dogs that develop persistent nasal discharge
Molecular Detection Using the Polymerase Chain Reaction (e.g., present for at least 1 week) should be encouraged to pursue
The use of real-time PCR assays that detect Aspergillus DNA in advanced imaging and rhinoscopy as soon as possible so that the
nasal biopsy specimens has been evaluated for diagnosis of SNA disease can be recognized and treated early. Treatment with anti-
in dogs, but assays evaluated had either unacceptably high rates bacterial drugs alone is discouraged, because primary bacterial
of false-positive test results (PenAsp assay) or a low sensitivity infections of the nasal cavity in adult dogs are rare.
(Aspergillus genus-specific PCR).34 For both assays, dogs with In general, treatment of SNA requires thorough mechanical
SNA had higher DNA copy numbers than control dogs or dogs debridement of plaques with endoscopic biopsy forceps until no
with lymphoplasmacytic rhinitis or neoplasia, but there was visible fungus remains, which may necessitate extended anes-
considerable overlap in copy numbers between dogs with SNA thesia. In some cases, treatment is performed immediately after
and those without SNA. imaging and rhinoscopy, which further prolongs the period of
anesthesia. If plaques within the frontal sinuses cannot be accessed
Pathologic Findings by antegrade rhinoscopy through the nasofrontal opening, fron-
Histopathology of nasal biopsies from dogs with SNA usually tal sinus trephination and curettage or open surgical debridement
reveals branching, septate fungal hyphae in a background of lym- of the frontal sinus may be required. Provided the cribriform
phoplasmacytic and neutrophilic inflammation; occasionally the plate is intact, debridement is followed by repeated lavage with
neutrophilic component predominates. Variable numbers of mac- 0.9% saline and suction, followed by topical antifungal drug
rophages and eosinophils may also be present, as well as necrotic therapy with clotrimazole or enilconazole. Additional systemic
debris. Other findings include osteolysis, turbinate remodeling, antifungal drug treatment could also be considered, but whether
and intralesional bacteria. Occasionally, intralesional plant mate- this improves outcome over topical therapy alone is unknown.
rial is detected. Mats of fungal hyphae may be present if a plaque
is submitted for examination. Conidiophores and conidia can be Topical Antifungal Drug Treatment
found because the fungus is exposed to air within the nasal cav- A variety of noninvasive and invasive protocols for topical anti-
ity (Figure 65-5). The sensitivity of rhinoscopically guided nasal fungal drug treatment of canine SNA have been described with
biopsy for diagnosis of canine SNA in one study was 82% (18 varied rates of success. Currently, there is a lack of masked,
of 22 dogs).25 In that study, three to five biopsy specimens were randomized clinical trials with consistent and long-term follow-
collected from each side of the nasal cavity. up evaluation that indicate clear benefit of one treatment over
Histopathology of biopsies of retrobulbar mass lesions in cats another. The protocol described here is used at the author’s
with SOA reveals granulomatous inflammation with necrosis and institution. A similar protocol has been used successfully to
intralesional branching hyphae. A mixed inflammatory reaction treat SNA in cats.38
CHAPTER 65  Aspergillosis 639

Of 14 dogs treated, 10 had resolution of clinical signs, but out-


Cribriform come was based only on telephone follow-up at 6 to 24 months,
plate
Infusion and not endoscopic reevaluation. In another study, 15 g of 1%
catheter bifonazole cream was administered into the frontal sinus using
Sponge endoscopic guidance either alone or after topical treatment with
Foley
enilconazole.24 Dogs were also treated with ketoconazole or
catheter itraconazole. All 12 dogs treated with a combined enilconazole-
bifonazole treatment were considered cured after a second treat-
ment, but 2 of 5 dogs treated with bifonazole alone had persistent
disease. Bifonazole has limited availability in the United States.

Endotracheal Soft palate Systemic Antifungal Drug Treatment


tube
Although their efficacy for treatment of SNA has been debated,
FIGURE 65-6  Diagram illustrating placement of catheters for topical treatment of azole antifungals with activity against molds (i.e., ketoconazole,
canine sinonasal aspergillosis with clotrimazole or enilconazole.
itraconazole, posaconazole, or voriconazole) have been used
either as monotherapy for dogs with cribriform plate involvement
A 24F Foley catheter is retroflexed over the soft palate and or in combination with topical antifungal drugs. Debridement
the balloon inflated until it can be palpated above the soft palate before commencing systemic antifungal drug treatment is recom-
(Figure 65-6). Placement of the catheter is facilitated by inser- mended if client finances permit, because effective drug concen-
tion of a stiff guidewire into the catheter. The tip of the cath- trations are not likely to be obtained within fungal plaques.
eter can then be bent into a U shape and hooked over the soft Cats with SOA must be treated with systemic antifungal
palate, after which the balloon is inflated. The nasopharynx is drugs such as an azole (itraconazole or posaconazole) and/or
packed with moistened laparotomy pads, a gauze pad is placed amphotericin B; the use of terbinafine in combination with these
over the incisive papilla, and the proximal end of the catheter drugs has also been reported.3 One cat was successfully treated
is clamped across the guidewire to prevent leakage of antifun- with caspofungin followed by posaconazole monotherapy.
gal drug from the nasal cavity. A 10F polypropylene catheter More than 50% of treated cats failed treatment in one study.3
is inserted into each nostril to the level of the medial canthus, The optimum treatment for SOA requires further study.
or alternatively these two catheters can be placed directly into
the frontal sinus with endoscopic guidance.24,39 The tip of an Prognosis
additional 12F Foley catheter is inserted into each of the nares, The prognosis for SNA depends on the extent of disease at the
the balloon inflated, and the catheters clamped so the nares time of diagnosis. Success rates vary from less than 30% to
are occluded. Cotton-tipped swabs can also be inserted into 100%, but methods to assess treatment success and follow-up
the nares to prevent outflow of antifungal drug. Clotrimazole times have not been consistent. Serial serologic testing by gel ID
(1%) or enilconazole (1% or 2%) solution is then administered is not useful to monitor treatment success.25 Even if infection is
through the polypropylene catheters. For large-breed dogs, 50 to eliminated based on endoscopic reevaluation and biopsy, dogs
60 mL of solution is administered per side, and if frontal sinus with extensive turbinate loss are predisposed to recurrent bacte-
trephination was performed, half of the solution can be adminis- rial infections of the nasal cavity, which result in persistent nasal
tered into a catheter placed in the trephination site. The patient discharge and sneezing. One study showed no difference in the
is rotated 45 degrees every 15 minutes over an hour to distribute prevalence of sneezing and nasal discharge between dogs that
the drug into the nasal cavity and sinuses. The catheters and responded to initial treatment and dogs that failed to respond.25
sponges are then removed with the dog’s head tilted downward Recurrence of SNA due to persistent infection or re-infection
at an angle of 30 degrees, and the material is allowed to drain by Aspergillus spp. can also occur. The prognosis for complete
from the nasal cavity for 10 minutes. Debridement and a second remission of SOA in cats is guarded to poor.
treatment are then routinely performed 1 month later; some-
times, a third treatment may be necessary. Clotrimazole can be Public Health Aspects
irritating to tissues, and solutions that contain propylene glycol
can cause pharyngeal irritation and edema.40 Enilconazole solu- There are no reports of transmission of SNA from affected dogs
tions may be less irritating, but there is no evidence that they are to humans. Whether the handling of affected dogs by immuno-
more effective than clotrimazole. suppressed humans poses a risk to their health is unknown, but
When cribriform plate destruction is present, topical treatment such individuals should probably minimize contact with affected
can lead to the development of life-threatening neurologic signs or animals. If contact is necessary, they should be encouraged to
failure to recover from anesthesia. Nevertheless, treatment has also discuss the problem with their medical practitioner. At the least,
been performed without complication in some dogs with minor gloves should be worn and proper hand washing practiced.
defects in the cribriform plate.25 Systemic azole antifungal drug
therapy could be considered as an alternative in this situation. Systemic Aspergillosis
Use of topical antifungal creams has been studied in order
to improve retention in the sinonasal cavities and reduce proce- Etiology and Epidemiology
dure time.24,41 In one study, 1% clotrimazole solution (25 or 60
mL based on patient size) was administered through a catheter Systemic aspergillosis may involve the respiratory tract (bron-
placed through a trephination site in each frontal sinus, followed chopulmonary aspergillosis) or may be a disseminated disease of
by a 1% clotrimazole cream (10 or 20 g).41 Debridement was multiple organ systems. Localized bronchopulmonary aspergil-
not performed, so the mean treatment time was only 32 minutes. losis is rare in dogs and cats, and often follows underlying local
640 SECTION 3  Fungal and Algal Diseases

or systemic immunodeficiency.6,42-44 German shepherds appear


to be predisposed to a form of bronchopulmonary aspergillosis TABLE 65-2
characterized by the development of cavitary lung lesions.6,45-47 Historical Signs in 30 Dogs with Disseminated Aspergillosis
Bronchopulmonary aspergillosis is typically associated with
infection by A. fumigatus complex fungi, whereas disseminated Sign Percent of Dogs
aspergillosis in dogs is most often caused by Aspergillus terreus
or Aspergillus deflectus. Other Aspergillus species have also been Inappetence or anorexia 20
isolated from dogs with disseminated disease (see Table 65-1). Pain 27
In dogs, disseminated aspergillosis may occur as a result of Lameness 13
an uncharacterized genetic immunodeficiency, because female
Neurologic signs (especially pelvic 20
German shepherds are strongly predisposed to the disease,
limb ataxia and paresis)
and immunosuppressive drug treatment is rarely present in the
history of affected dogs.7 Two thirds of affected dogs seen at Respiratory signs (especially cough) 13
the author’s institution were German shepherds, and German Vomiting  7
shepherds were 43-fold more likely to develop the disease than
other dog breeds.7 Rhodesian ridgebacks were also predisposed. From Schultz RM, Johnson EG, Wisner ER, et al. Clinicopathologic and
Females are 3 times as likely to develop disseminated aspergil- diagnostic imaging characteristics of systemic aspergillosis in 30 dogs.
J Vet Intern Med. 2008;22:851-859.
losis as males, and the median age of affected dogs is 4.5 years
(range, 2 to 8 years). Concurrent or sequential infections with
other opportunistic pathogens have been described in some
dogs.4,48
Disseminated and bronchopulmonary aspergillosis are rare TABLE 65-3
in cats.42,49,50 Affected cats often have recognizable underlying
immunosuppressive conditions such as diabetes mellitus, cancer
Physical Examination Findings in 30 Dogs with Disseminated
chemotherapy, or severe viral infections. The remainder of this Aspergillosis
chapter focuses on systemic aspergillosis in dogs.
Finding Percent of Dogs
Clinical Features Muscle wasting/thin body condition 40
Fever 27
Signs and Their Pathogenesis
Spinal pain 17
Pulmonary or disseminated aspergillosis is thought to develop
after inhalation of Aspergillus conidia, which lodge in the alve- Vestibular signs 17
oli and adhere to epithelial cells. In the absence of adequate Peripheral lymphadenomegaly 17
local immune defenses, the conidia enlarge and germinate to Ocular abnormalities 13
form hyphae. Invasion of the bloodstream and dissemination to
distant sites may occur as a result of systemic immunosuppres- Lameness 13
sion. Fungal virulence factors are also likely to be important, Ataxia 10
although why A. terreus and A. deflectus are more likely than Paraparesis 10
A. fumigatus to cause disseminated disease in dogs is unclear. Cough, increased breath sounds 10
Human disseminated aspergillosis is most often caused by
A. fumigatus–complex fungi.51 Mental dullness  7
Anatomic sites of predilection in dogs with disseminated Vision impairment  7
aspergillosis are the vertebral endplates and discs, renal pelvis, Hemiparesis  7
spleen, long bones, and lymph nodes. However, any organ may
Circling, seizures, arrhythmia  3
be affected, including the liver, heart, cardiac valves, pancreas,
eyes, lungs, brain, bone marrow, meninges, small intestine, From Schultz RM, Johnson EG, Wisner ER, et al. Clinicopathologic and
skin, spinal cord, thyroid, adrenal glands, prostate, and trachea. diagnostic imaging characteristics of systemic aspergillosis in 30 dogs.
Nasal cavity involvement is not generally a feature of the bron- J Vet Intern Med. 2008;22:851-859.
chopulmonary or disseminated forms, and dogs with dissemi-
nated disease often lack evidence of pulmonary involvement.
The median duration of illness before admission in one study with disseminated aspergillosis often have muscle wasting and/
was 1 month (range, 2 days to 9 months).7 The most frequent or a thin body condition. Fever (up to 104.2°F or 40.1°C) is
clinical signs are lethargy, inappetence, pain, lameness, and present in one quarter of affected dogs.7 Musculoskeletal abnor-
neurologic signs secondary to discospondylitis (especially ataxia malities include spinal pain, lameness, and, rarely, inability to
and paresis) (Table 65-2). Other signs include apparent blind- stand. Neurologic signs include vestibular abnormalities, ataxia,
ness, circling, head tilt, seizures, respiratory distress, and vomit- mental dullness, paraparesis, vision impairment, hemiparesis,
ing. Cough, increased respiratory effort, and rarely hemoptysis circling, and seizures. Dogs with respiratory involvement may
may occur in dogs with bronchopulmonary aspergillosis.6,47 cough and have harsh lung sounds and/or increased respiratory
effort. Peripheral lymphadenomegaly may be detected. Ocular
Physical Examination Findings abnormalities include chorioretinitis, hyphema, and panophthal-
Physical examination findings in dogs with disseminated asper- mitis.7 Rarely, arrhythmias are detected secondary to myocardial
gillosis are listed in Table 65-3. On physical examination, dogs involvement, but myocardial involvement is often subclinical.
CHAPTER 65  Aspergillosis 641

TABLE 65-4
Selected Hematologic and Serum Biochemistry Analysis Results in 30 Dogs with Disseminated Aspergillosis
Median
Variable (Range) No. (%) with Low Values No. (%) with High Values Reference Range
Hematocrit (%) 42% 8 (31%) 0 40%-55%
(14%-46%)
Neutrophils 12,643 0 21 (81%) 3000-10,500
(cells/µL) (7584-28,321)
Band neutrophils 0 0 10 (38%) Rare
(cells/µL) (0-3051)
Monocytes 1608 0 16 (62%) 150-1200
(cells/µL) (499-3760)
Lymphocytes 1566 4 (15%) 0 1000-4000
(cells/µL) (115-3998)
Platelets 262,000 1 (4%) 0 150,000-400,000
(cells/µL) (86,000-400,000)
Albumin (g/dL) 2.7 6 (24%) 0 2.9-4.2
(1.3-3.6)
Globulin (g/dL) 4.4 0 12 (48%) 2.3-4.4
(2.3-5.8)
Creatinine (g/dL) 1.1 0 9 (36%) 0.5-1.6
(0.5-7.8)
BUN (mg/dL) 19 0 9 (36%) 8-31
(8-196)
Calcium (mg/dL) 10.8 0 8 (32%) 9.9-11.4
(9.9-13.9)
ALT (IU/L) 37 0 3 (12%) 19-67
(19-609)
ALP (IU/L) 78 0 3 (12%) 15-127
(21-1920)

Diagnosis Cerebrospinal Fluid Analysis


Analysis of the CSF in dogs with neurologic signs or spinal pain
Laboratory Abnormalities due to disseminated aspergillosis can show no abnormalities,
Complete Blood Count or an increased nucleated cell count and CSF protein concen-
Most dogs with disseminated aspergillosis have leukocyto- tration. The differential cell count reveals mixed or primarily
sis due to a neutrophilia (Table 65-4).7 A left shift and toxic mononuclear or neutrophilic pleocytosis. Abnormalities of the
neutrophils may also be present. Normocytic, normochromic CSF were identified in 8 of 10 dogs with disseminated aspergil-
nonregenerative anemia occurs in approximately one third of losis at the author’s teaching hospital.4
affected dogs and may be secondary to inflammatory disease or
renal failure. Diagnostic Imaging
Plain Radiography
Serum Biochemical Tests Plain thoracic radiographs in dogs with bronchopulmonary
Common abnormalities in the serum biochemical profiles of aspergillosis may show a bronchial pattern or lung lobe consoli-
dogs with systemic aspergillosis include hyperglobulinemia, azo- dation.44 Consolidation and cavitary lesions may be seen in the
temia, hypercalcemia, and hypoalbuminemia (see Table 65-4). lungs of German shepherd dogs (Figure 65-7).45-47 Abnormali-
ties in dogs with disseminated aspergillosis include enlarged tra-
Urinalysis cheobronchial and/or sternal lymph nodes, cranial mediastinal
The urine specific gravity of azotemic dogs with disseminated masses, pleural effusion, and/or pulmonary alveolar infiltrates.7
aspergillosis is usually in the isosthenuric range because of Spinal radiographs reveals evidence of discospondylitis in
renal failure. Hematuria, proteinuria, and pyuria may also be approximately half of dogs with disseminated aspergillosis, with
detected. Fungal hyphae were identified in the urine sediment a median of 2 sites affected (range, 1 to 9 sites).7 Productive and
of 2 (8%) of 25 dogs with disseminated aspergillosis in one destructive bony changes consistent with osteomyelitis may be
study.7 present in other parts of the skeleton (Figure 65-8). Most dogs
642 SECTION 3  Fungal and Algal Diseases

A B
FIGURE 65-7  Lateral thoracic radiograph of a 2-year-old male neutered German shepherd that had a 4-month history of cough and exercise intolerance. A, A cavitary lesion (arrows)
and alveolar infiltrates in the right middle lung lobe are present. B, On a horizontal beam projection, a fluid line is present (arrow) within the dependent aspect of the lesion.

FIGURE 65-9  Ultrasound image of the right kidney of a 6-year-old female spayed
German shepherd dog with disseminated aspergillosis. There is dilation of the renal pelvis,
which contains echogenic material (arrow).

abdominal lymph nodes.7 Renal abnormalities include pyelecta-


sia, a distorted and mottled architecture, decreased corticome-
dullary distinction, echogenic debris within the renal pelvis and
proximal ureter, nodules or masses within the renal cortex, and
papillary blunting (Figure 65-9). Occasionally, hydronephrosis
is detected. Splenic abnormalities include hypoechoic nodules or
masses, splenomegaly, changes consistent with infarction, or a
mottled echotexture. Abdominal lymph nodes may be enlarged
and/or hypoechoic. Other findings include diffuse hepatic or
pancreatic hypoechogenicity, ascites, or evidence of venous
FIGURE 65-8  Lateral radiograph of the left humerus of a 2-year-old male neutered thrombosis.
German shepherd with disseminated aspergillosis. A primarily osteoproductive lesion is
present in the mid-diaphysis.
Advanced Imaging
MRI of the head in dogs with Aspergillus infections that have
have multiple affected sites, which include the vertebral bodies, disseminated to the central nervous system (CNS) may reveal
sternebrae, ribs, scapula, humerus, and tibia.7 multifocal lesions that are hypo- to isointense and enhance with
contrast on T1-weighted images. These lesions are hyperintense
Sonographic Findings or heterogenous on T2-weighted and fluid attenuated inversion
Abdominal ultrasound abnormalities in dogs with dissemi- recovery (FLAIR) images. Mild or severe meningeal enhance-
nated aspergillosis are most often in the kidneys, spleen, and ment may also be present.
CHAPTER 65  Aspergillosis 643

TABLE 65-5
Diagnostic Assays Available for Aspergillosis in Dogs or Cats
Assay Specimen Type Target Performance
Cytologic Aspirates of affected Fungal hyphae False negatives can occur when specimen size is small or low
­examination tissues, body fluids, (with or without numbers of fungi are present. Cytology of brush specimens
impression smears of conidia in tissues optimizes sensitivity for dogs with SNA. ­Identification to
biopsies exposed to air) species level requires culture and ­molecular testing.
Fungal culture Aspirates or biopsies of Aspergillus species Sensitivity for diagnosis of canine SNA is at least 75% when
­affected tissues, body endoscopically guided nasal biopsies are cultured. Specific-
fluids ity is >95%. Culture of a typical species
(A. terreus or A. deflectus) from a dog with signs ­consistent
with disseminated disease strongly suggests disseminated
aspergillosis. Molecular testing by PCR may be required to
identify beyond the species complex level.
Antibody Serum Antibodies to Assays vary in sensitivity and specificity. In general, positive
­serology (gel ­Aspergillus species results support a diagnosis of canine SNA, but negative
ID or ELISA) results do not rule out the disease. Insensitive for diagnosis
of disseminated aspergillosis.
Galactomannan Urine, serum Aspergillus spp. Insensitive for diagnosis of SNA, SOA, and probably
antigen assay ­galactomannan bronchopulmonary disease. Very sensitive for diagnosis
(ELISA) cell wall antigen of disseminated aspergillosis. Cross-reactivity with other
fungal pathogens may occur (especially other molds). False
positives also occur with Plasmalyte administration.
Real-time PCR Whole blood Aspergillus spp. Assays studied lack sensitivity or specificity for diagnosis
assays DNA of canine SNA. Usefulness for diagnosis of disseminated
aspergillosis requires further study.
Histopathology Biopsy specimens from Fungal hyphae Sensitivity for diagnosis of canine SNA in rhinoscopically
­affected tissues (with or without guided biopsies was 82%. Hyphae are usually visualized in
conidia in tissues biopsies from animals with disseminated aspergillosis, but
exposed to air) lesions may be difficult to access for biopsy. Special stains
assist organism detection.

Microbiologic Tests cytologic evidence of fungal hyphae is required for definitive


Diagnostic assays available for aspergillosis in dogs or cats are diagnosis in this situation.
described in Table 65-5. Once growth of mold occurs in culture, Aspergillus species
can be identified to the complex level based on conidiophore
Cytologic Examination morphology (Figure 65-10). Accurate identification to the
Hyphae with morphology consistent with Aspergillus spp. may be species level requires molecular techniques such as PCR, then
present in aspirates of lymph nodes, kidney, lung tissue, or bone sequencing, which are performed by laboratories with special
from dogs with systemic aspergillosis (see Figure 4-3).7 Cytologic expertise (e.g., the Fungus Testing Laboratory at the University
examination of pleural effusion, airway lavage specimens, or of Texas Health Science Center at San Antonio). Such labora-
urine may also reveal hyphae. False-negative results occur when tories can also perform antifungal drug susceptibility testing.
organism numbers are low or specimen size is small. Accurate However, because clinical breakpoints have not been estab-
identification of the organism present requires fungal culture. lished for dogs or cats, the relevance of minimum inhibitory
concentrations for molds remains unclear.
Fungal Culture
Aspergillus spp. are readily isolated on routine laboratory Serologic Diagnosis
media. Isolation of Aspergillus spp. (especially A. terreus or An Aspergillus galactomannan antigen ELISA assay (Platelia)
A. deflectus) from normally sterile sites such as urine, blood, has high sensitivity for diagnosis of disseminated aspergillosis in
or aspirates of lymph node, bone, intervertebral disc, liver, or dogs when serum or urine specimens are tested.36 In the author’s
splenic tissue supports a diagnosis of disseminated aspergillo- hospital, all of 12 dogs with disseminated aspergillosis had posi-
sis. Approximately 50% of dogs with disseminated aspergillosis tive test results, with galactomannan indices (GMIs) that ranged
have positive urine cultures for Aspergillus spp., and around from 6.4 to 12.2 (reference range, <0.5). Two dogs with local-
one third of dogs have positive blood or CSF cultures.7 Isola- ized pulmonary disease had serum GMIs of 0.14 and 0.77, which
tion of A. fumigatus from airway washes should be interpreted ­suggests that, as for SNA, the sensitivity of this assay for diag-
more cautiously as it may represent contamination. Concurrent nosis of bronchopulmonary aspergillosis may be low. High-level
644 SECTION 3  Fungal and Algal Diseases

A B
FIGURE 65-10  A, Morphology of Aspergillus fumigatus. B, Morphology of Aspergillus terreus, which has an “upswept” appearance. The conidiophore is the specialized hyphal branch
that bears the conidia (spores). The vesicle is the dilated top part of the conidiophore. Phialides are flask-shaped projections from the vesicle from which the conidia arise.

false positive test results (GMIs >1.5) appear to only occur in pale, tan to white masses or nodules within a variety of paren-
dogs with other fungal infections, especially those caused by chymal organs or the pleura (Figure 65-11, A); evidence of
Penicillium or Paecilomyces, and dogs treated with Plasmalyte discospondylitis and/or osteomyelitis; serosanguineous pleu-
intravenous fluid therapy. Plasmalyte contains sodium gluconate, ral effusion; and/or mottled and congested lung lobes. Splenic
which is produced by fermentation in ­Aspergillus niger; carry- infarction is also a common finding. The renal pelvis may be
over of small amounts of galactomannan during production of dilated and contain soft, granular material. Histopathology
the fermentation product is thought to occur.52 Low-level false reveals granulomatous or pyogranulomatous inflammation
positives (0.5-1.5) can occur in dogs infected with other fungi with abundant intralesional fungal hyphae in a variety of tis-
(such as Cryptococcus spp.), some dogs with nonfungal disease sues. Some species, such as A. terreus, also produce accessory
such as neoplasia, or some dogs treated with penicillin deriva- conidia (aleuriospores) in tissues, which may be free or extend
tives. In summary, a strong positive assay result in a dog with from hyphae. Fibrinoid necrosis of blood vessels and associated
clinical abnormalities consistent with disseminated aspergillosis hemorrhage may be present. Special stains such as Gomori’s
and no history of Plasmalyte administration within the last 72 methenamine silver or periodic acid–Schiff can delineate fungal
hours supports a diagnosis of a mold infection, but fungal culture hyphae in tissues (Figure 65-11, B). Immunohistochemistry has
is required to confirm that Aspergillus is the causative agent. A also been used to identify Aspergillus.55,56
negative result in a dog with disseminated disease suggests that
Aspergillus is not the cause, but it does not rule out the possibility Treatment and Prognosis
of infection with another mold species.
Assays that detect antibodies to Aspergillus species are insen- Many dogs with disseminated aspergillosis have advanced disease
sitive for diagnosis of systemic aspergillosis.7 This may reflect at the time of evaluation, sometimes with respiratory distress,
underlying host immunodeficiency (and hence inability to produce pathologic fractures, and vertebral subluxations and cord com-
antibodies), or antibodies in affected dogs may not cross-react pression. The prognosis for these dogs is poor. In one study, 57%
with the antigens used in the test kits. For example, one widely of 30 dogs were euthanized within a week of examination (median,
used gel ID assay (Meridian Bioscience) contains a purified carbo- 3 days).7 However, dogs that lack severe CNS signs, pain, or respi-
hydrate preparation from mycelial phase cultures of A. fumigatus, ratory distress may enter remission for months and occasionally
A. niger, and A. flavus, but not A. terreus or A. deflectus. years when treated with antifungal drugs. These dogs maintain
high antigen titers or persistently positive urine cultures, and they
Molecular Diagnosis Using the Polymerase Chain Reaction ultimately relapse and die from the disease. Sometimes, euthanasia
Several real-time PCR assays have been developed and applied is performed earlier because continued treatment is unaffordable.
for the diagnosis of invasive aspergillosis in humans.53,54 The The relative efficacy of different antifungal drug treatment
sensitivity and specificity of these assays has varied.54 To date, regimens for disseminated aspergillosis in dogs has not been
they have not been widely applied to diagnosis of disseminated studied prospectively. The most widely used treatments have
aspergillosis in dogs. been itraconazole, voriconazole, posaconazole, and/or ampho-
tericin B. Terbinafine has been used in addition to azole therapy
Pathologic Findings with limited success in the author’s experience.7 Aspergillus spe-
Gross pathologic findings in dogs with disseminated aspergillo- cies are intrinsically resistant to fluconazole, so the use of fluco-
sis include enlarged and/or congested lymph nodes; multifocal, nazole is not recommended. Although expensive, treatment with
CHAPTER 65  Aspergillosis 645

FIGURE 65-12  Severe ulcerative keratitis in a 12-year-old female spayed domestic


shorthair caused by an Aspergillus fumigatus complex organism. The condition was suc-
cessfully treated using topical voriconazole and a conjunctival island graft. (Courtesy the
University of California, Davis, Veterinary Ophthalmology Service.)

for successful treatment of German shepherds with cavitary


lung lesions.7,45,46

Immunity and Vaccination


Immunity to disseminated aspergillosis is critically dependent
on the function of neutrophils and dendritic cells, as well as
effective T cell function.62 A vaccine to prevent the disease is
not available.
B
FIGURE 65-11  A, Kidney at necropsy from a 5-year-old female English setter with Public Health Aspects
disseminated aspergillosis caused by Aspergillus terreus. Numerous small coalescing tan
nodules are present throughout the cortex, and the capsular surface is irregular. There is In human patients, invasive aspergillosis primarily occurs in
a blood clot adjacent to the renal pelvis. (Image courtesy University of California, Davis, immunosuppressed humans with hematologic malignancy, or
Veterinary Anatomic Pathology Service.) B, Histopathology from a 5-year-old male neu- after solid organ transplantation or hematopoietic stem cell
tered German shepherd dog with disseminated A. terreus infection that involved multiple transplantation.51 Infections are acquired from the environment.
parenchymal organs. Large numbers of septate, branching hyphae are seen. Gomori’s Transmission from affected dogs has not been described. Most
methenamine silver stain, 1000× oil magnification.
cases (>70%) are caused by A. fumigatus. Infection with A. ­terreus
is rare, and A. deflectus is not a recognized human pathogen.
voriconazole or posaconazole can result in remissions of many
months’ duration in some dogs.7,57 Posaconazole in particular KERATOMYCOSIS AND OTOMYCOSIS
has shown promising activity in human patient and mouse mod- CAUSED BY ASPERGILLUS SPECIES
els of invasive A. terreus infection.58,59 Many dogs require treat-
ment with multiple antifungal drugs to maintain remission, either Infection of the cornea8,63 and ear canal9,10 by Aspergillus spe-
in combination or in series. A. terreus strains from humans with cies occur rarely in dogs and cats (see Table 65-1; Figure 65-12).
invasive aspergillosis can become resistant to azoles during treat- Diagnosis of these conditions requires visualization of hyphae on
ment.60 A. terreus strains isolated from human patients are also cytologic examination or histopathology of biopsy specimens in
relatively resistant to amphotericin B,59,61 but clinical improve- addition to fungal culture to rule out the possibility of contami-
ment can still occur after treatment of infected dogs with ampho- nation or colonization. One cat with A. flavus keratomycosis had
tericin B (see Chapter 9).7 Echinocandins (such as caspofungin) a history of feline herpesvirus-1–associated conjunctivitis and
are also active against Aspergillus, but expense and the need for keratitis8 and was successfully treated with 1% topical voricon-
intravenous administration has limited their use in dogs. azole (q2h to q4h). Cats with Aspergillus otomycosis often have
Dogs with localized bronchopulmonary aspergillosis have a history of diabetes mellitus.4 Dogs with otomycosis may have
the best prognosis. These dogs are sometimes cured after treat- a history of underlying allergic dermatitis and otitis externa that
ment with itraconazole alone. Lung lobectomy may be required has been extensively treated with topical and oral antibiotics.10
646 SECTION 3  Fungal and Algal Diseases

CASE EXAMPLE Albumin 2.8 g/dL (2.9-4.2 g/dL)


Globulin 4.0 g/dL (2.3-4.4 g/dL)
ALT 15 U/L (19-67 U/L)
Signalment: “Zach,” a 2-year-old male neutered German AST 27 U/L (21-54 U/L)
shepherd from Roseville in northern California ALP 28 U/L (15-127 U/L)
History: Zach was evaluated at the University of California, GGT 2 U/L (0-6 U/L)
Davis, Veterinary Oncology Service for possible bone cancer. Cholesterol 205 mg/dL (135-345 mg/dL)
Two months previously, the owners noted that Zach vocalized Total bilirubin 0.1 mg/dL (0-0.4 mg/dL).
when he stood up from rest. He was taken to a local veterinary Urinalysis: SGr 1.010; pH 7.0, 1+ protein (SSA), no bilirubin, 3+
clinic where he was diagnosed with lumbosacral pain and hemoprotein, no glucose, 10-15 WBC/HPF, 50-60 RBC/HPF,
treated with carprofen (1.5 mg/kg PO q12h). Two weeks later, 0-2 transitional epithelial cells, a few amorphous crystals,
right pelvic limb lameness developed. Pelvic radiographs and a few budding organisms were seen.
showed evidence of moderate osteoarthrosis of the left Imaging Findings:
coxofemoral joint. Carprofen treatment was continued. After Thoracic Radiographs: The cardiopulmonary structures were
an additional 2 weeks, lethargy, inappetence, increased thirst unremarkable.
and urination, and left thoracic limb lameness developed, Bone Survey Radiographs: Single lateral radiographs of
and Zach began vomiting once daily. Radiographs of the the distal left antebrachium, distal right antebrachium,
left thoracic limb revealed an osteoproductive lesion in the and distal left and right pelvic limbs were performed. An
mid-diaphysis of the left humerus, which was interpreted as additional lateral radiograph of the left humerus was also
a primary or metastatic bone tumor (see Figure 65-8). Zach obtained and compared with referral radiographs taken 1
was one of two dogs in the household, was normally fed a dry month previously. This showed progression of a primarily
commercial dog food diet, and was up to date on vaccinations, osteoproductive lesion in the mid-diaphysis of the left
which included those for distemper, adenovirus, parvovirus, humerus. An additional site of chronic active periosteal
rabies, Borrelia, and Bordetella. reaction was noted along the distal caudal aspect of the left
Physical Examination: femur. Patchy increases in medullary density were identified
Body Weight: 33.5 kg. in the distal diaphysis of both humeri.
General: Quiet, alert, and responsive. T = 103.4°F (39.7°C), HR Abdominal Ultrasound: Hypoechoic mottling of the splenic
= 120 beats/min, panting, CRT <2 s, mucous membranes hilus was identified. There was bilateral renal pyelectasia
moist and pink. with echogenic material within the renal pelves bilaterally
Eyes, Ears, Nose, and Throat: No clinically significant (see Figure 65-9). There was marked blunting of the renal
abnormalities were detected. Fundoscopic examination pelves bilaterally with mild hydroureter.
revealed multiple hyporeflective regions, consistent with Microbiologic Findings: Cytologic examination of a fine needle
chorioretinitis, in the right fundus. aspirate of lesion in left humerus: Smears were moderately
Musculoskeletal: BCS was 4/9, and generalized muscle cellular with a dense, proteinaceous background and
wasting was present. There was no evidence of lameness or moderate amounts of cellular debris. Nucleated cells consisted
bone pain on orthopedic examination. of a population of numerous activated macrophages and
Cardiovascular, Respiratory, Gastrointestinal, Genitouri- variably degenerate neutrophils that were evenly distributed
nary, and Lymph Nodes: No clinically significant abnor- throughout the smear as well as in variably sized aggregates.
malities were present. Numerous multinucleated giant cells were noted. Throughout
Laboratory Findings: the specimen were moderate numbers of small, round, yeast
CBC: organisms as well as numerous fungal hyphae, which were
HCT 32.8% (40%-55%) found intracellularly and extracellularly. The fungal hyphae were
MCV 65.2 fL (65-75 fL) negative staining, branching, and septate, often with bulging
MCHC 35.7 g/dL (33-36 g/dL) ends.
WBC 18,530 cells/µL (6000-13,000 cells/µL) Fungal culture and susceptibility (left humerus fine needle aspi-
Neutrophils 13,768 cells/µL (3000-10,500 cells/µL) rate, sent to University of Texas for molecular identification):
Lymphocytes 2687 cells/µL (1000-4000 cells/µL) Moderate numbers of Aspergillus terreus. MICs for ampho-
Monocytes 1334 cells/µL (150-1200 cells/µL) tericin B, itraconazole, and voriconazole were 2, 0.125, and
Eosinophils 741 cells/µL (0-1500 cells/µL) 0.25 µg/mL, respectively.
Platelets 262,000 platelets/µL (150,000-400,000 platelets/µL). Aerobic urine bacterial culture: Six colonies of A. terreus.
Serum Chemistry Profile: Aspergillus antibody serology (gel immunodiffusion, Meridian
Sodium 152 mmol/L (145-154 mmol/L) Bioscience, Ltd.): Negative.
Potassium 3.8 mmol/L (4.1-5.3 mmol/L) Diagnosis: Disseminated Aspergillus terreus infection.
Chloride 113 mmol/L (105-116 mmol/L) Treatment: Zach was treated with intravenous fluids (lactated
Bicarbonate 24 mmol/L (16-26 mmol/L) Ringer’s solution with 20 mEq/L KCl at 75 mL/hr), lipid-
Phosphorus 5.7 mg/dL (3.0-6.2 mg/dL) complexed amphotericin B (1 mg/kg IV on a Monday-
Calcium 11.8 mg/dL (9.9-11.4 mg/dL) Wednesday-Friday basis for 4 weeks), and itraconazole
BUN 31 mg/dL (8-21 mg/dL) (5 mg/kg PO q12h). Over the course of amphotericin B
Creatinine 1.9 mg/dL (0.5-1.6 mg/dL) treatment, Zach’s inappetence, lethargy, and lameness
Glucose 107 mg/dL (60-104 mg/dL) resolved and his serum creatinine and BUN concentrations
Total protein 6.8 g/dL (5.4-7.4 g/dL) remained stable with values of 2.1 mg/dL and 49 mg/dL,
CHAPTER 65  Aspergillosis 647

respectively, by the end of treatment. At a recheck 2 months of the masses revealed suppurative inflammation and possible
after treatment was initiated, fungal culture of the urine occasional fungal hyphae. Bronchoscopy was performed, and
revealed a single colony of A. terreus, and the creatinine was 1.7 numerous yellow plaque-like lesions were visualized on the
mg/dL. After an additional month, bone survey radiographs tracheal mucosa. Cytologic examination of biopsies of these
showed significant remodeling of the left humeral and left lesions revealed fungal hyphae. The owner elected euthana-
femoral lesions. A serum biochemistry panel showed mildly sia. Necropsy findings included severe, locally extensive mural
increased creatinine concentration (1.8 mg/dL) and a serum granulomas of the tracheal mucosa with intralesional fungal
ALT activity of 364 U/L; the increased serum ALT activity hyphae and suppurative and eosinophilic pharyngitis with
was thought to be secondary to itraconazole administration. necrosis and edema. There was evidence of healed pyelone-
Ultrasound of the abdomen showed mild pelvic dilatation, phritis and granulomatous nephritis with intralesional fungal
and the echogenic material within the renal pelves had hyphae. Granulomatous osteomyelitis with intralesional hy-
disappeared. Fungal culture of the urine was negative. Four phae was present in the left femur and humerus. There were
months after treatment was initiated, intermittent vocalization multiple granulomas in the pancreas and spleen.
and right pelvic limb lameness developed. Pain was detected Comments: Disseminated aspergillosis in this dog was initially
only on lumbosacral palpation. Radiographs of this region confused with bone neoplasia. An Aspergillus antibody titer
revealed focal osteolysis of the ventral caudal aspect of the was negative, which illustrates the lack of sensitivity of this
sixth lumbar vertebra. Clinical improvement did not occur assay for diagnosis of disseminated disease. Galactomannan
after treatment with carprofen and tramadol. antigen testing was not performed, but bone lesions were
Two weeks later, Zach developed respiratory distress and was readily accessible for aspiration and culture. The small, round
seen by the Veterinary Emergency and Critical Care Service. yeast organisms visualized together with the hyphae were
Mild dehydration, pyrexia (105.8°F or 41.0°C), gagging, and likely accessory conidia, which can be found in A. terreus
ptyalism were detected on physical examination. Thoracic infections. Treatment with lipid-complexed amphotericin
radiographs were unremarkable, but a soft tissue density was B and itraconazole was initially associated with clinical
seen in the pharyngeal region on cervical radiographs. Blood remission, but infection was not eliminated. Based on
cultures were negative. Zach was sedated, and on oral exami- necropsy examination, the cause of the suppurative
nation, multiple large soft tissue masses were observed within pharyngeal masses that impaired respiration was not clear.
the pharynx and around the rim of the epiglottis. Aspiration Fungal cultures of the masses were negative.

SUGGESTED READINGS 10. Coyner K. Otomycosis due to Aspergillus spp. in a dog: case report
and literature review. Vet Dermatol. 2010;21:613-618.
Barrs VR, Halliday C, Martin P, et al. Sinonasal and sino-orbital asper- 11. Zhang S, Corapi W, Quist E, et  al. Aspergillus versicolor, a new
gillosis in 23 cats: aetiology, clinicopathological features and treat- causative agent of canine disseminated aspergillosis. J Clin Micro-
ment outcomes. Vet J. 2012;191:58-64. biol. 2012;50:187-191.
Day MJ. Canine sino-nasal aspergillosis: parallels with human disease. 12. Barrs VR, van Doorn T, Houbraken J, et  al. Aspergillus felis sp.
Med Mycol. 2009;47(suppl 1):S315-S323. nov. – an emerging pathogen of cats, dogs and humans. Proceed-
Schultz RM, Johnson EG, Wisner ER, et al. Clinicopathologic and diag- ings of the International Society for Companion Animal Infectious
nostic imaging characteristics of systemic aspergillosis in 30 dogs. Diseases. 2012: Abstract 017.
J Vet Intern Med. 2008;22:851-859. 13. Talbot JJ, Martin P, Johnson LR, et  al. What causes sinonasal
aspergillosis in dogs? A molecular approach to species identifica-
REFERENCES tion. Proceedings of the International Society for Companion Ani-
1. Denning DW. Invasive aspergillosis. Clin Infect Dis. 1998;26:781- mal Infectious Diseases. 2012: Abstract 018.
803:quiz 804-785. 14. Burrough E, Deitz K, Kinyon J, et al. Disseminated aspergillosis in
2. Abad A, Fernandez-Molina JV, Bikandi J, et al. What makes Asper- a dog due to Aspergillus alabamensis. Med Mycol Case Reports.
gillus fumigatus a successful pathogen? Genes and molecules involved 2012;1:1-4.
in invasive aspergillosis. Rev Iberoam Micol. 2010;27:155-182. 15. Pitt JI, Samson RA. Nomenclatural considerations in naming spe-
3. Barrs VR, Halliday C, Martin P, et al. Sinonasal and sino-orbital cies of Aspergillus and its teleomorphs. Stud Mycol. 2007;59:67-70.
aspergillosis in 23 cats: aetiology, clinicopathological features and 16. Pomrantz JS, Johnson LR, Nelson RW, et al. Comparison of sero-
treatment outcomes. Vet J. 2012;191:58-64. logic evaluation via agar gel immunodiffusion and fungal culture
4. Sykes JE. Unpublished observations, 2012. of tissue for diagnosis of nasal aspergillosis in dogs. J Am Vet Med
5. Kano R, Itamoto K, Okuda M, et al. Isolation of Aspergillus uda- Assoc. 2007;230:1319-1323.
gawae from a fatal case of feline orbital aspergillosis. Mycoses. 17. Day MJ. Canine sino-nasal aspergillosis: parallels with human dis-
2008;51:360-361. ease. Med Mycol. 2009;47(suppl 1):S315-S323.
6. Southard C. Bronchopulmonary aspergillosis in a dog. J Am Vet 18. Goodall SA, Lane JG, Warnock DW. The diagnosis and treat-
Med Assoc. 1987;190:875-877. ment of a case of nasal aspergillosis in a cat. J Small Anim Pract.
7. Schultz RM, Johnson EG, Wisner ER, et al. Clinicopathologic and 1984;25:627-633.
diagnostic imaging characteristics of systemic aspergillosis in 30 19. Peeters D, Day MJ, Clercx C. An immunohistochemical study of
dogs. J Vet Intern Med. 2008;22:851-859. canine nasal aspergillosis. J Comp Pathol. 2005;132:283-288.
8. Labelle AL, Hamor RE, Barger AM, et al. Aspergillus flavus kera- 20. Peeters D, Peters IR, Clercx C, et  al. Quantification of mRNA
tomycosis in a cat treated with topical 1% voriconazole solution. encoding cytokines and chemokines in nasal biopsies from dogs
Vet Ophthalmol. 2009;12:48-52. with sino-nasal aspergillosis. Vet Microbiol. 2006;114:318-326.
9. Ghibaudo G, Peano A. Chronic monolateral otomycosis in a dog 21. Hamilton HL, Whitley RD, McLaughlin SA. Exophthalmos second-
caused by Aspergillus ochraceus. Vet Dermatol. 2010;21:522-526. ary to aspergillosis in a cat. J Am Anim Hosp Assoc. 2000;36:343-347.
648 SECTION 3  Fungal and Algal Diseases

22. Giordano C, Gianella P, Bo S, et al. Invasive mould infections of the 42. Hazell KL, Swift IM, Sullivan N. Successful treatment of pulmo-
naso-orbital region of cats: a case involving Aspergillus fumigatus nary aspergillosis in a cat. Aust Vet J. 2011;89:101-104.
and an aetiological review. J Feline Med Surg. 2010;12:714-723. 43. Sykes JE, Mapes S, Lindsay LL, et  al. Corynebacterium ulcerans
23. Barachetti L, Mortellaro CM, Di Giancamillo M, et  al. Bilat- bronchopneumonia in a dog. J Vet Intern Med. 2010;24:973-976.
eral orbital and nasal aspergillosis in a cat. Vet Ophthalmol. 44. Adamama-Moraitou KK, Pardali D, Day MJ, et  al. Aspergillus
2009;12:176-182. fumigatus bronchopneumonia in a Hellenic shepherd dog. J Am
24. Billen F, Guieu LV, Bernaerts F, et  al. Efficacy of intrasinusal Anim Hosp Assoc. 2011;47:e13-e18.
administration of bifonazole cream alone or in combination with 45. Kulendra E, Halfacree Z, Goggs R, et  al. Cavitary pulmonary
enilconazole irrigation in canine sino-nasal aspergillosis: 17 cases. lesion associated with Aspergillus fumigatus infection in a German
Can Vet J. 2010;51:164-168. shepherd dog. J Small Anim Pract. 2010;51:271-274.
25. Pomrantz JS, Johnson LR. Repeated rhinoscopic and serologic 46. Whitley NT, Cauvin A, Burton C, et al. Long term survival in two
assessment of the effectiveness of intranasally administered clotrim- German shepherd dogs with Aspergillus-associated cavitary pulmo-
azole for the treatment of nasal aspergillosis in dogs. J Am Vet Med nary lesions. J Small Anim Pract. 2010;51:561.
Assoc. 2010;236:757-762. 47. Guerin SR, Walker MC, Kelly DF. Cavitating mycotic pneumonia
26. Saunders JH, Clercx C, Snaps FR, et  al. Radiographic, magnetic in a German shepherd dog. J Small Anim Pract. 1993;34:36-39.
resonance imaging, computed tomographic, and rhinoscopic 48. Krockenberger MB, Swinney G, Martin P, et al. Sequential oppor-
features of nasal aspergillosis in dogs. J Am Vet Med Assoc. tunistic infections in two German Shepherd dogs. Aust Vet J.
2004;225:1703-1712. 2011;89:9-14.
27. Johnson LR, Drazenovich TL, Herrera MA, et  al. Results of rhi- 49. Fox JG, Murphy JC, Shalev M. Systemic fungal infections in cats.
noscopy alone or in conjunction with sinuscopy in dogs with J Am Vet Med Assoc. 1978;173:1191-1195.
aspergillosis: 46 cases (2001-2004). J Am Vet Med Assoc. 50. Ossent P. Systemic aspergillosis and mucormycosis in 23 cats. Vet
2006;228:738-742. Rec. 1987;120:330-333.
28. Whitney BL, Broussard J, Stefanacci JD. Four cats with fungal rhi- 51. Steinbach WJ, Marr KA, Anaissie EJ, et al. Clinical epidemiology
nitis. J Feline Med Surg. 2005;7:53-58. of 960 patients with invasive aspergillosis from the PATH alliance
29. Peeters D, Clercx C. Update on canine sinonasal aspergillosis. Vet registry. J Infect. 2012;65:453-464.
Clin North Am Small Anim Pract. 2007;37:901-916, vi. 52. Hage CA, Reynolds JM, Durkin M, et al. Plasmalyte as a cause of
30. Smith LN, Hoffman SB. A case series of unilateral orbital aspergil- false-positive results for Aspergillus galactomannan in bronchoal-
losis in three cats and treatment with voriconazole. Vet Ophthal- veolar lavage fluid. J Clin Microbiol. 2007;45:676-677.
mol. 2010;13:190-203. 53. White PL, Linton CJ, Perry MD, et  al. The evolution and evalu-
31. De Lorenzi D, Bonfanti U, Masserdotti C, et  al. Diagnosis of ation of a whole blood polymerase chain reaction assay for the
canine nasal aspergillosis by cytological examination: a compari- detection of invasive aspergillosis in hematology patients in a rou-
son of four different collection techniques. J Small Anim Pract. tine clinical setting. Clin Infect Dis. 2006;42:479-486.
2006;47:316-319. 54. Johnson GL, Bibby DF, Wong S, et al. A MIQE-compliant real-time
32. Billen F, Clercx C, Le Garerres A, et al. Effect of sampling method PCR assay for Aspergillus detection. PLoS One. 2012;7:e40022.
and incubation temperature on fungal culture in canine sinonasal 55. Butterworth SJ, Barr FJ, Pearson GR, et al. Multiple discospondy-
aspergillosis. J Small Anim Pract. 2009;50:67-72. litis associated with Aspergillus species infection in a dog. Vet Rec.
33. Billen F, Peeters D, Peters IR, et  al. Comparison of the value of 1995;136:38-41.
measurement of serum galactomannan and Aspergillus-specific 56. Perez J, Mozos E, de Lara FC, et  al. Disseminated aspergillo-
antibodies in the diagnosis of canine sino-nasal aspergillosis. Vet sis in a dog: an immunohistochemical study. J Comp Pathol.
Microbiol. 2009;133:358-365. 1996;115:191-196.
34. Peeters D, Peters IR, Helps CR, et al. Whole blood and tissue fungal 57. Legendre AM. Unpublished observations, 2012.
DNA quantification in the diagnosis of canine sino-nasal aspergil- 58. Salas V, Pastor FJ, Rodriguez MM, et  al. In  vitro activity and
losis. Vet Microbiol. 2008;128:194-203. in vivo efficacy of posaconazole in treatment of murine infections
35. Tomsa K, Glaus TM, Zimmer C, et al. Fungal rhinitis and sinusitis by different isolates of the Aspergillus terreus complex. Antimicrob
in three cats. J Am Vet Med Assoc. 2003;222:1380-1384, 1365. Agents Chemother. 2011;55:676-679.
36. Garcia RS, Wheat LJ, Cook AK, et al. Sensitivity and specificity of 59. Hachem RY, Kontoyiannis DP, Boktour MR, et  al. Aspergil-
a blood and urine galactomannan antigen assay for diagnosis of lus terreus: an emerging amphotericin B–resistant opportunis-
systemic aspergillosis in dogs. J Vet Intern Med. 2012;26:911-919. tic mold in patients with hematologic malignancies. Cancer.
37. Whitney J, Beatty JA, Martin P, et al. Evaluation of serum galac- 2004;101:1594-1600.
tomannan detection for diagnosis of feline upper respiratory tract 60. Arendrup MC, Jensen RH, Grif K, et  al. In  vivo emergence of
aspergillosis. Vet Microbiol. 2013;162:180-185. Aspergillus terreus with reduced azole susceptibility and a Cyp51a
38. Furrow E, Groman RP. Intranasal infusion of clotrimazole for the M217I alteration. J Infect Dis. 2012;206:981-985.
treatment of nasal aspergillosis in two cats. J Am Vet Med Assoc. 61. Escribano P, Pelaez T, Recio S, et  al. Characterization of clinical
2009;235:1188-1193. strains of Aspergillus terreus complex: molecular identification and
39. McCullough SM, McKiernan BC, Grodsky BS. Endoscopically antifungal susceptibility to azoles and amphotericin B. Clin Micro-
placed tubes for administration of enilconazole for treatment of biol Infect. 2012;18:E24-E26.
nasal aspergillosis in dogs. J Am Vet Med Assoc. 1998;212:67-69. 62. Park SJ, Burdick MD, Mehrad B. Neutrophils mediate maturation
40. Caulkett N, Lew L, Fries C. Upper-airway obstruction and pro- and efflux of lung dendritic cells in response to Aspergillus fumiga-
longed recovery from anesthesia following intranasal clotrimazole tus germ tubes. Infect Immun. 2012;80:1759-1765.
administration. J Am Anim Hosp Assoc. 1997;33:264-267. 63. Qualls Jr CW, Chandler FW, Kaplan W, et al. Mycotic keratitis in a
41. Sissener TR, Bacon NJ, Friend E, et  al. Combined clotrimazole dog: concurrent Aspergillus sp. and Curvularia sp. infections. J Am
irrigation and depot therapy for canine nasal aspergillosis. J Small Vet Med Assoc. 1985;186:975-976.
Anim Pract. 2006;47:312-315.
CHAPTER 66

Rhinosporidiosis
Jane E. Sykes

majority of affected dogs are young-adult to middle-aged (with


Overview of Rhinosporidiosis a range of 1 to 13 years of age), otherwise-healthy, large-breed
First Described: In the 1890s, in Argentina (by Malbran and hunting dogs such as Labrador and golden retrievers, Doberman
then by Seeber)1 pinschers, Siberian huskies, German shepherds, and Rhodesian
ridgebacks. However, small-breed dogs can also be affected.7
Cause: Rhinosporidium seeberi (kingdom Protista, class
No clear sex predisposition has been identified, but of cases
Mesomycetozoea)
in the literature, males have outnumbered females. Affected
Affected Hosts: Dogs, humans, horses, rarely cats and other cats have been outdoor cats. One affected cat had concurrent
domestic animals nasal adenocarcinoma,8 but other cats have had no reported
Geographic Distribution: Worldwide but especially in warm, comorbidities.
wet environments such as the south-eastern and south- Although the mode of transmission of R. seeberi is not pre-
central United States cisely understood, disease often occurs in association with con-
tact with aquatic or marshy environments. It is thought that
Mode of Transmission: Unclear; possibly exposure to contam- spores are released into the environment, where they contact
inated water sources in association with trauma susceptible human and animal hosts.2 Spores may be introduced
Major Clinical Signs: Sneezing, epistaxis, mass protruding into tissues after trauma to the mucous membranes, where they
from nares form sporangia that produce new spores. Because disease in
Differential Diagnoses: Nasal neoplasia, cryptococcosis, sino- humans also occurs in arid regions, airborne spore transmission
nasal aspergillosis, nasal mites (Pneumonyssoides cani- has also been suggested.
num), foreign bodies
Clinical Features
Human Health Significance: Human infections by R. seeberi
are likely acquired from the environment, and direct Signs and Their Pathogenesis
transmission from affected animals to humans has not In dogs and cats, infection by R. seeberi leads to formation of
been described. pedunculate or sessile masses within the nasal cavity, usually
in the rostral third.6 The polyps range in size from a few mil-
limeters to several centimeters. Clinical signs consist of sub-
acute (weeks) to chronic (>1 year) sneezing with or without
Etiology and Epidemiology epistaxis, snuffling, and sometimes a serous, mucoid, or sero-
sanguineous unilateral nasal discharge. Sneezing may be fre-
Rhinosporidiosis is caused by Rhinosporidium seeberi, which quent and severe and associated with excitement or increased
causes slow-growing tumor-like masses in the nasal cavity.
Although once thought to be a fungus, molecular methods
have identified this organism as an aquatic protistan parasite
(class Mesomycetozoea). The organism is taxonomically located
where animals and fungi diverge and is closely related to patho-
gens of fish.2 The organism is difficult or impossible to culture.
Rhinosporidiosis has been most widely reported in humans and
dogs, but cats, horses, and other mammalian and avian species
may also be affected. There is some evidence that host-specific
strains of R. seeberi exist.3
In humans, rhinosporidiosis is most prevalent in southern
India, Sri Lanka, and Argentina but also occurs in North Amer-
ica, Africa, Europe, and Asia. In dogs and cats, rhinosporidi-
osis has been reported from many regions of the United States,
Ontario in Canada,4 Argentina,5 Italy in continental Europe,6
and the United Kingdom.7 Within the United States, the disease
is most often reported in dogs and cats from the south- FIGURE 66-1  States within the United States in which rhinosporidiosis has been
­central and southeastern states (Figure 66-1). The overwhelming reported in dogs and cats.

649
650 SECTION 3  Fungal and Algal Diseases

FIGURE 66-2  CT scan of the nasal cavity of a 16-month-old female Labrador retriever FIGURE 66-3  Rhinoscopic image of a polypoid mass caused by Rhinosporidium see-
with rhinosporidiosis. A soft tissue mass that enhanced with contrast is present in the beri in the nasal cavity of a 10-year-old intact female yellow Labrador retriever.
­rostral nasal cavity (arrow).

activity. In some dogs, a polypoid pink mass protrudes from


the nasal cavity. The masses are often stippled with white-
yellowish granules less than 1 mm in diameter, which represent
mature sporangia.6

Diagnosis
Most animals with rhinosporidiosis have no significant abnor-
malities on blood work. Plain radiography is insensitive for
detection of nasal masses caused by R. seeberi.6 Computed
tomography (CT) reveals soft tissue masses in the rostral third
of the nasal cavity that can enhance with contrast (Figure 66-2).
In some cases evidence of turbinate loss is present. The masses,
which can be visualized directly using rostral rhinoscopy, are FIGURE 66-4  Impression smear of a biopsy taken from the nasal cavity of a 6-year-
pink to gray and may be covered with miliary white to yellow old female spayed Labrador retriever with rhinosporidiosis. A large cluster of endospores is
foci, but these are not always clearly visible (Figure 66-3). present together with several epithelial cells.
Definitive diagnosis requires cytology or histopathology
of the masses. Cytologic examination of smears from nasal
swabs, cytology brush specimens, or impression smears made be seen releasing endospores at the polyp surface through a pore
from biopsies of the polyps reveals suppurative to pyogranu- in the sporangial wall. Although the organisms stain well with
lomatous inflammation with dysplastic epithelial cells and special stains such as periodic acid–Schiff, Gomori’s methena-
numerous immature to mature R. seeberi endospores, which mine silver, and Mayer’s mucicarmine, these stains are usually
occur singly or in clusters.9 Sometimes sporangia that contain not required for diagnosis.6
hundreds of endospores are visualized. Mature endospores
stain intensely, are round to oval and 10 to 15 µm in diam- Treatment and Prognosis
eter, and have a thick cell wall (Figure 66-4). Immature endo-
spores are 2 to 4 µm in diameter and contain a paracentral, The treatment of choice for dogs and cats is surgical removal
light pink-purple structure that may represent nuclear mate- of polyps, which generally requires rhinotomy. Because the
rial, as well as one to two smaller, spherical, dark purple polyps are often rostrally located, excisions can often be
structures.9 extended caudally from the nares. Many dogs have no recur-
Histopathology reveals abundant sporangia in various stages rence of clinical signs thereafter, but some dogs develop
of development and a mild to severe pyogranulomatous inflam- relapse within months of surgery.5,10,11 Treatment of these
matory response (Figure 66-5). Lymphocytes, plasma cells, and, dogs can be frustrating and costly. Povidone-iodine is active
in some cases, multinucleate giant cells and evidence of turbi- against R. seeberi in  vitro and could be applied in the form
nate remodeling with activated osteoclasts and osteoblasts may of gauze packs topically after surgery in an attempt to mini-
be present. Juvenile sporangia are small (<100 µm in diameter) mize the chance of recurrence (see Case Example).12 In some
and have a single nucleus that is surrounded by fibrillar mate- dogs, apparent transient or long-term responses to treatment
rial. Mature sporangia are up to 500 µm in diameter and may with ketoconazole7,13 or dapsone (1.1 mg/kg PO q8-12h)11
CHAPTER 66  Rhinosporidiosis 651

A B
FIGURE 66-5  Histopathology of a polyp caused by Rhinosporidium seeberi. A, Low-power image that shows sporangia in various stages of development. B, High-power image
(1000× oil magnification) that shows a juvenile sporangium adjacent to a mature sporangium, which contains endospores (right). H&E stain.

have been described. Responses to dapsone have also been Public Health Aspects
described in human patients. Dapsone could be used to treat
dogs with refractory disease, but because both dapsone and In humans, rhinosporidiosis usually involves the nasal cavity
ketoconazole can have significant adverse effects, careful mon- or nasopharynx, but involvement of the conjunctiva, genita-
itoring is required. Dapsone is a sulfur drug (diaminophenyl lia, or skin can occur.15 Rarely, cutaneous disease is dissemi-
sulfone) that can cause bone marrow suppression in dogs14 nated. Disease is most often reported in males. Infection is likely
and has the potential to cause hepatotoxicity and cutaneous acquired from the environment, and transmission from animals
drug reactions. Treatment of cats with dapsone is not recom- to humans has not been described. It is possible that strains of
mended because of a high prevalence of adverse effects in this R. seeberi that infect humans may differ from those that infect
species, specifically neurotoxicosis and anemia. dogs.

CASE EXAMPLE Physical Examination:


Body Weight: 32.2 kg.
General: Excited, alert, responsive, and hydrated. T = 101.6°F
Signalment: “Sarah,” a 16-month-old intact female Labrador (38.7°C), HR = 140 beats/min, panting, CRT <2 s, mucous
retriever from the central valley of northern California membranes moist and pink.
History: Sarah was evaluated by her local veterinary clinic for Eyes, Ears, Nose, and Throat: The dog’s face was symmetrical
a 1-month history of sneezing, which occurred throughout with no pain on palpation, and normal ocular retropulsion.
the day and was more frequent and severe during periods There was no evidence of nasal discharge. Nasal airflow
of excitement. The owners had not observed nasal was absent on the right side. Minimal dental calculus was
discharge, and they were concerned about the possibility of present. There were no other clinically significant findings.
a foreign body. A limited rhinoscopic examination with an Musculoskeletal: BCS 4/9. Adequate musculature, normal gait.
otoscope cone at the local veterinary clinic revealed a large, Cardiovascular, Respiratory, Gastrointestinal, Genitourinary,
fleshy, irregular mass in the dorsal meatus near the nares. and Lymph Nodes: No clinically significant abnormalities
Histopathology on a biopsy of the mass yielded a diagnosis were detected.
of rhinosporidiosis, and Sarah was referred to the University Laboratory Findings: Preanesthetic laboratory assessment:
of California, Davis, Veterinary Medical Teaching Hospital for PCV 44%, total solids 6.6 g/dL. Serum urea nitrogen was
further evaluation and treatment. estimated at 15-26 mg/dL with a reagent strip.
Sarah had been obtained from a breeder in Oregon as a puppy Imaging Findings: Computed tomography of the head: A
and since then had been a duck hunting dog in California. round, 0.9 × 1.7 × 2.3 cm, soft tissue mass was present in the
During the hunting season, she waded and swam in stag- rostral, dorsal right nasal cavity extending from the level of
nant and fresh water sources. The owners also had ponds in the incisors to the root of the right maxillary canine tooth.
their backyard. She was otherwise healthy and was fed a dry The mass was moderately and heterogeneously contrast
commercial dog food diet. enhancing. The superficial soft tissues overlying the mass

Continued
652 SECTION 3  Fungal and Algal Diseases

were thickened and contrast enhancing. The remainder of The dog was discharged with instructions to administer
the nasal cavities and sinuses appeared within normal limits. tramadol (3 mg/kg PO q12h) for pain relief and dapsone
The mandibular and medial retropharyngeal lymph nodes (0.8 mg/kg PO q8h for 2 weeks, then 0.8 mg/kg PO q12h
appeared within normal limits. for 3 months). At a recheck 2 weeks later, the sneezing had
Diagnosis: Nasal rhinosporidiosis. reduced in frequency, but vomiting was reported every
Treatment: A rhinotomy was performed using an anterolateral 2 days approximately 2 hours after the morning dose of
approach, and a single 1.5 cm × 0.5 cm pedunculated mass dapsone. A CBC and biochemistry panel were unremarkable.
was identified attached to the mucosal layer of the nasal The frequency of dapsone administration was reduced to
septum. The mass was resected with sharp dissection and twice daily. Subsequently Sarah did well with no recurrence
diathermy. A povidone-iodine–soaked gauze pad was of disease or adverse effects of medication.
placed on the site of resection for 7 minutes, and the nasal Comments: In this dog, topical povidone-iodine and dapsone
vestibule was packed with an iodine-soaked gauze pad for were used after rhinotomy in an attempt to prevent
1 hour after surgical closure. Histopathology of the resected recurrence. The owners were warned about the possibility
mass confirmed the diagnosis of nasal rhinosporidiosis. of reinfection if Sarah continued her hunting activities.

SUGGESTED READING 7. Miller RI, Baylis R. Rhinosporidiosis in a dog native to the UK. Vet
Rec. 2009;164:210.
Caniatti M, Roccabianca P, Scanziani E, et al. Nasal rhinosporidiosis in 8. Brenseke BM, Saunders GK. Concurrent nasal adenocarcinoma
dogs: four cases from Europe and a review of the literature. Vet Rec. and rhinosporidiosis in a cat. J Vet Diagn Invest. 2010;22:155-157.
1998;142:334-338. 9. Meier WA, Meinkoth JH, Brunker J, et al. Cytologic identification
of immature endospores in a dog with rhinosporidiosis. Vet Clin
REFERENCES Pathol. 2006;35:348-352.
1. Fredricks DN, Jolley JA, Lepp PW, et al. Rhinosporidium seeberi: a 10. Mosier DA, Creed JE. Rhinosporidiosis in a dog. J Am Vet Med
human pathogen from a novel group of aquatic protistan parasites. Assoc. 1984;185:1009-1010.
Emerg Infect Dis. 2000;6:273-282. 11. Allison N, Willard MD, Bentinck-Smith J, et  al. Nasal rhinospo-
2. Mendoza L, Vilela R, Rosa PS, et  al. Lacazia loboi and Rhino- ridiosis in two dogs. J Am Vet Med Assoc. 1986;188:869-871.
sporidium seeberi: a genomic perspective. Rev Iberoam Micol. 12. Arseculeratne SN, Atapattu DN, Balasooriya P, et al. The effects of
2005;22:213-216. biocides (antiseptics and disinfectants) on the endospores of Rhino-
3. Silva V, Pereira CN, Ajello L, et al. Molecular evidence for multiple sporidium seeberi. Indian J Med Microbiol. 2006;24:85-91.
host-specific strains in the genus Rhinosporidium. J Clin Microbiol. 13. Easley JR, Meuten DJ, Levy MG, et al. Nasal rhinosporidiosis in
2005;43:1865-1868. the dog. Vet Pathol. 1986;23:50-56.
4. Hoff B, Hall DA. Rhinosporidiosis in a dog. Can Vet J. 1986;27: 14. Lees GE, McKeever PJ, Ruth GR. Fatal thrombocytopenic hemor-
231-232. rhagic diathesis associated with dapsone administration to a dog.
5. Castellano MC, Idiart JR, Martin AA. Rhinosporidiosis in a dog. J Am Vet Med Assoc. 1979;175:49-52.
Vet Med Small Anim Clinician. 1984;79:45. 15. Prasad K, Veena S, Permi HS, et al. Disseminated cutaneous rhino-
6. Caniatti M, Roccabianca P, Scanziani E, et al. Nasal rhinosporidi- sporidiosis. J Lab Physicians. 2010;2:44-46.
osis in dogs: four cases from Europe and a review of the literature.
Vet Rec. 1998;142:334-338.
CHAPTER 67

Candidiasis
Jane E. Sykes

Overview of Candidiasis BOX 67-1


First Described: The etiology of candidiasis as a fungus was
Disease Syndromes Caused by Candida in Dogs and Cats
recognized in Stockholm around 1840 (Fredrik Berg),1 but
the disease was described as far back as around 400 bc by
Hippocrates. Keratitis
Cutaneous candidiasis
Cause: Candida spp. (an ascomycete) Otitis externa and otitis media
Affected Hosts: Dogs, cats, humans, and a variety of other Urinary tract infections
animal species Gastrointestinal candidiasis
Peritonitis
Geographic Distribution: Worldwide
Disseminated infections
Mode of Transmission: Usually invasion of commensal yeasts
secondary to immune suppression
Major Clinical Signs: Local dermatitis; otitis externa; keratitis;
lower urinary tract signs; abdominal pain, icterus, and/or When compared with human patients, candidiasis is
ascites due to peritonitis; or severe systemic infections uncommonly described in dogs and cats. Predisposing factors
with fever, inappetence, ocular inflammation, lameness in dogs or cats with local or disseminated candidiasis have
due to osteomyelitis, respiratory distress, vomiting included diabetes mellitus,2,3,12 treatment with immunosup-
Differential Diagnoses: Other systemic fungal or bacterial pressive drugs or broad-spectrum antibacterial drugs,2,13 a his-
infections tory of gastrointestinal surgery,4,5 parvoviral infections,14-16
Human Health Significance: Human infections by Candida and/or underlying malignancy.2,17 Often more than one of
species result from invasion by normal flora, but human- these factors is in the history. Disseminated candidiasis has
to-human transmission of Candida strains can also occur. most often been described in dogs, but also occurs in cats.5,18
Whether resistant Candida species can be transferred from Although most animals have underlying immunocompromis-
animals to colonize humans or vice versa is not known. ing conditions, disseminated candidiasis has been described
in apparently immunocompetent animals,19,20 which may
reflect an unidentified underlying genetic immunodeficiency.
Cats with candidiasis usually test negative for FeLV or FIV
Etiology and Epidemiology infection.
Analysis of the author’s hospital population suggests that
Candida species are yeasts that are typically benign counterparts cats are at least as likely as dogs to develop Candida UTIs.11
of the normal gastrointestinal, urogenital, and cutaneous flora In other studies, Candida species were isolated from the urine
but can invade tissues and cause disease as a result of disrup- of 0.2% of more than 8000 dogs with UTIs21 and 0.2% of
tion of normal host defenses. Candida species can also be iso- more than 2000 dogs with persistent or recurrent UTIs.22 Ure-
lated from soil, inanimate objects, and hospital environments. throstomy or indwelling cystostomy tube placement may be in
The yeasts are small (3 to 6 µm) and ovoid (blastospores) and the history of some cats and dogs with Candida UTIs, respec-
reproduce by budding. Budding results in the formation of new tively.2,6 Local mucocutaneous infections can occur at the site of
yeast cells, pseudohyphae (chains of elongated yeast cells), and long-term placement of feeding tubes.7
true septate hyphae. Disease syndromes caused by Candida spe-
cies that occur in dogs and cats are listed in Box 67-1. More than Clinical Features
200 species of Candida exist, but only a few species have been
identified as pathogens in dogs and cats (Table 67-12-10). The Signs and Their Pathogenesis
most commonly isolated species is Candida albicans, and only Disruption of normal barriers is necessary to allow Candida spp.
C. albicans appears to cause disseminated disease in dogs and to invade the host. Concurrent use of broad-spectrum antibiot-
cats. Local or disseminated infections where Candida is present ics suppresses the normal bacterial flora and allows Candida to
with a mixture of bacterial species can occur. Local infections proliferate, which increases the risk that organisms will be intro-
(such as UTIs) with two different Candida species have also been duced into tissues. Candida can adhere to a variety of tissue com-
reported.2,11 ponents such as epithelial and endothelial cells, fibronectin, and

653
654 SECTION 3  Fungal and Algal Diseases

thrombi, as well as other bacteria and inanimate objects such as in the immune response to Candida.24 The formation of hyphae
catheters.23 The yeasts can also form biofilms on medical devices by Candida promotes tissue invasion, which may be followed by
and produce a huge variety of hydrolytic enzymes. Within tis- life-threatening fungemia and dissemination to multiple organs.
sues, neutrophils are a key defense against Candida.24 Candida This ability of Candida to change its morphology (“morphologic
blastospores are phagocytized and destroyed by neutrophils. dimorphism”) is thought to be an important virulence factor.
Mononuclear cells play a less important role. Reactive oxygen Lower urinary tract candidiasis may be subclinical or asso-
species, hydrolytic enzymes, and antimicrobial peptides contrib- ciated with dysuria, stranguria, and/or hematuria. Systemic
ute to the intracellular destruction of blastospores by neutro- signs of pyrexia, lethargy, inappetence, and weight loss have
phils. Thus, any condition that impairs neutrophil numbers or also been described in affected dogs and cats,6 but these signs
function, such as neutropenia or diabetes mellitus, predisposes may result from the presence of other predisposing diseases.
dogs and cats to candidiasis. A large number of other cytokines, Ascending infections may lead to development of Candida
complement, dendritic cells, and lymphocytes are also involved pyelonephritis.
Cutaneous candidiasis may manifest as erythema, scaling,
alopecia, erosions, and crusting, often where disruption of
cutaneous or mucocutaneous regions has occurred (such as a
TABLE 67-1 previous surgical site or a feeding tube or cystotomy stoma).
Candida Species Associated with Disease in Dogs and/or Cats 2-10 Persistent disease in the face of systemic or topical treatment
with broad-spectrum antibiotics should raise suspicion for can-
Species Clinical Manifestations didiasis. Dogs with Candida otitis externa often have a history
of chronic bacterial otitis externa that has been treated aggres-
Candida albicans Dermatitis, UTI, otitis externa, sively with systemic or topical antibacterials.
keratitis, disseminated infections Overgrowth of Candida in the gastrointestinal tract may occur
Candida glabrata (formerly UTI, feeding-tube site infections in animals that are immunosuppressed or treated with glucocor-
Torulopsis glabrata) ticoids and/or broad-spectrum antibacterial drugs. In some cases,
Candida krusei UTI, feeding tube site infections invasion of the mucosa into the lamina propria or deeper tissues
occurs (e.g., intestinal candidiasis). Intestinal candidiasis is occa-
Candida guilliermondii UTI, dermatitis
sionally identified at necropsy in puppies that die after treatment
Candida parapsilosis UTI, otitis, dermatitis for parvoviral enteritis and may be a reason for persistent vomit-
Candida tropicalis UTI ing, diarrhea, and death despite aggressive fluid and antibacterial
Candida rugosa UTI drug therapy.14 Candida spp. may also contribute to ulcerative
glossitis in puppies with parvovirus infection (Figure 67-1).

A B
FIGURE 67-1  A, Tongue ulcerations at necropsy in a 2-month-old mixed-breed puppy that was euthanized because of canine parvoviral infection. B, Histopathology revealed mul-
tifocal ulcerative glossitis, pharyngitis, and esophagitis with intralesional yeasts and pseudohyphae. Gomori’s methenamine silver stain. (Image courtesy University of California Davis
Anatomic Pathology Service.)
CHAPTER 67  Candidiasis 655

Candida peritonitis can follow intestinal surgery, treatment Grossly, urine may contain flocculent white debris. Urinalysis
with broad-spectrum antibacterial drugs, and subsequent enter- may reveal proteinuria, pyuria, and, in some cases, hematuria
otomy site dehiscence.4,5 Mixed infections with Candida and and/or Candida fungal elements (see Case Example). Dogs and
intestinal flora may develop. Affected dogs are febrile, have cats with disseminated candidiasis or Candida peritonitis fre-
abdominal pain, and may have other gastrointestinal signs such quently have had nonregenerative anemia and mild to marked
as vomiting and diarrhea. neutrophilia with bandemia and toxic neutrophil changes (in
Disseminated candidiasis results from hematogenous contrast to other deep mycoses, where hematologic abnormali-
spread of Candida from intestinal, urinary, or possibly corneal ties are often mild or absent). Lymphopenia may be present as a
or cutaneous sites.12,15,25A diagnosis of disseminated candi- result of underlying immunosuppressive illness or drug therapy.
diasis should be considered whenever Candida infections are Some dogs are thrombocytopenic and have coagulation abnor-
identified in the eye, skin, or urinary tract of an animal that malities consistent with disseminated intravascular coagula-
has (or subsequently develops) severe systemic illness. Clini- tion.3 Findings on the serum biochemistry panel in dogs with
cal signs of disseminated candidiasis in dogs include fever, disseminated candidiasis are variable and nonspecific but may
inappetence, anorexia, weight loss, and a variety of other include metabolic acidosis, moderate to severe hypoalbumin-
clinical signs that reflect underlying immunosuppressive dis- emia, and increased liver enzyme activities.
ease processes and specific organs involved. The latter can
include the pancreas, liver, mesentery, spleen, kidneys, heart, Diagnostic Imaging
lungs, lymph nodes, and, less often, the bone, intestinal tract, Plain thoracic radiographs in dogs with disseminated candidia-
eyes, meninges, thyroid, and/or prostate gland.3,12,15,17,20,25- sis may be unremarkable or show increased pulmonary inter-
28 Ocular signs in animals with disseminated disease include stitial opacity, thoracic lymphadenomegaly, and/or pleural or
keratitis and corneal ulceration, uveitis, chorioretinitis, and pericardial effusion.13,5,20 Abdominal ultrasound examination
endophthalmitis.11,12,28 Candida keratitis may also occur in dogs with Candida peritonitis or disseminated disease may
as a localized process, sometimes secondary to aggressive reveal echogenic ascites fluid, mesenteric hyperechogenicity,
treatment of corneal ulceration secondary to other disease changes consistent with pancreatitis, abdominal lymphadeno-
processes with topical broad-spectrum antibacterial drugs. megaly, or increased echogenicity of the renal cortices or hepatic
Thromboembolic disease may complicate disseminated infec- parenchyma.
tions. Pulmonary thromboembolism may result in respiratory
distress and death.3,11 Microbiologic Tests
Definitive diagnosis of candidiasis requires visualization of the
Diagnosis organism within lesions by cytologic or histopathologic exami-
nation and confirmation of its identity using culture.
Laboratory Abnormalities
Laboratory abnormalities in dogs and cats with candidiasis Cytologic Examination
result from the underlying disease process (such as diabetes Candida blastospores, pseudohyphae, and true hyphae can be
mellitus or systemic neoplastic disease) as well as the location visualized in swab specimens from the ear canal (ear ­cytology);
and severity of the fungal infection. Laboratory abnormalities tape preparations from the skin; urine sediment or other
in dogs or cats with Candida UTIs may be absent, or there may body fluids (Figure 67-2); or ultrasound-guided aspirates of
be evidence of renal failure with azotemia and isosthenuria. ­abdominal lesions.

A B
FIGURE 67-2  Candida albicans within urine sediment from a 5-year-old female spayed Labrador retriever being treated for multicentric lymphoma and acute-onset hematuria. Both
budding (A) and filamentous (B) fungal elements are visible. (Image courtesy Dr. Barrak Pressler, The Ohio State University. From Bartges J, Polzin D. Nephrology and Urology of Small
Animals. John Wiley and Sons, 2011.)
656 SECTION 3  Fungal and Algal Diseases

Fungal Culture urine, such as fluconazole with or without 5-flucytosine. Sys-


Candida grows on routine bacteriologic media as well as fungal temic antifungals alone may be adequate to resolve Candida
media such as Sabouraud dextrose agar. Creamy white colo- UTIs in some animals, although resolution has been reported
nies appear within 2 to 3 days, and so isolation of the yeast in some animals in the absence of antifungal drug therapy.2
may be reported when it is present in specimens submitted for Persistent infection may result from drug resistance, adverse
aerobic bacterial culture, which are typically incubated only for effects of treatment that necessitate treatment discontinuation,
this period of time. If Candida is suspected based on the his- or inadequate drug penetration into balls of fungus that line
tory, fungal culture (rather than bacterial culture alone) should the bladder wall (see Case Example). Intravesicular treatment
be requested because Candida grows best on fungal isolation with 1% clotrimazole may be effective in these cases.30,31 Under
media. Because Candida can be isolated from the skin and anesthesia, the bladder is catheterized with a Foley catheter,
mucous membranes of healthy dogs and cats, infection must emptied, then lavaged with sterile saline. The bladder is then
be confirmed with cytology or histopathology when Candida is filled with 1% clotrimazole solution (e.g., 30 to 50 mL for a
isolated from these sites. cat). The animal is rotated to promote contact of the solution
Identification of Candida isolates is based on morphology with the uroepithelium, and after the solution has been in the
and biochemical reactions. Preliminary differentiation of C. bladder for an hour, the catheter is removed and the animal is
albicans from other Candida species can be made based on allowed to recover. Treatment is continued weekly until Can-
the ability of C. albicans to produce germ tubes when exposed dida can no longer be isolated from the urine. Typically one to
to serum. Matrix-assisted laser desorption/ionization–time of four treatments are required.11,30 Ultrasound-guided adminis-
flight (MALDI-TOF) mass spectrometry (see Chapter 3) accu- tration of clotrimazole via cystocentesis to a dog with Candida
rately and rapidly identifies Candida species29 and may replace cystitis was described in one report.31
these methods in the future. Isolation of Candida allows not Successful treatment of systemic candidiasis in dogs or cats is
only definitive identification but also antifungal susceptibility rarely described. One dog with Candida peritonitis was treated
testing (see Figure 4-8). This is important for Candida because successfully by surgical exploration and lavage of the abdomen,
some isolates (especially C. glabrata and C. krusei) can exhibit placement of an abdominal drain, and intravenous fluconazole.4
antifungal drug resistance. In addition, aggressive supportive measures were required that
included intravenous fluid therapy, fresh frozen plasma, antiemet-
Pathologic Findings ics, gastroprotectants, total parenteral nutrition, and antibacterial
Gross pathologic findings at necropsy in animals with dissemi- drugs for a suspected catheter-related infection. Antifungal drugs
nated candidiasis include peritoneal or pleural effusion, enlarged other than fluconazole used to treat invasive candidiasis in human
and edematous lymph nodes, congestion and necrotic whitish patients include amphotericin B or an echinocandin such as
foci within affected organs, evidence of tissue thrombosis, and caspofungin,32 both of which must be administered parenterally.
formation of a gray-white or yellow-green fibrinonecrotic layer
on affected mucosal surfaces.3,5,14,15,20,26 Concurrent underlying Prevention
disease processes are also often identified.
Histopathology of affected tissues reveals yeasts, pseudo- Prevention of candidiasis involves avoidance of excessive immu-
hyphae, and septate hyphae with an associated inflammatory nosuppression as well as discriminate use of antibacterial drugs,
response that is composed primarily of neutrophils, macro- which should be tailored to the results of culture and susceptibil-
phages, and lymphocytes. Multinucleated giant cells may also ity whenever possible. Whether Candida species carried on the
be present.15,20,26 Pseudohyphae and hyphae predominate in hands of humans can colonize dogs and cats is not known, but
invasive infections (see Figure 4-1). Evidence of blood vessel routine hand washing and the wearing of gloves when immu-
invasion, thrombosing vasculitis, and infarction may be seen, nosuppressed animals are handled or intravascular devices are
with multifocal mycotic granulomas in affected organs.15 The placed should minimize the chance of health care–associated
organism is most readily visualized with special stains such infections. Although prophylactic antifungal drug treatment has
as periodic acid–Schiff and Gomori’s methenamine silver. been used to prevent invasive candidiasis in human high-risk
Immunohistochemistry has also been used to identify Candida groups, the low incidence of candidiasis in immunosuppressed
within lesions.15,20 When mixed infections with Candida and dogs and cats does not warrant the use of prophylactic antifungal
other bacteria are present, tissue gram stains may reveal bac- drugs in this group.
terial rods and/or cocci in addition to gram-positive fungal
elements. Public Health Aspects
Treatment and Prognosis Candida species (especially C. albicans) are common causes
of mucosal disease in humans. Since 2000, Candida has also
Treatment of candidiasis should involve management of under- emerged as an extremely important cause of invasive infections
lying immunosuppressive disorders, if possible, and specific owing to the more widespread use of invasive medical devices
antifungal drug treatment. Systemic antibacterial drugs should and immunosuppressive drugs. In hospitalized human patients,
be discontinued if not clearly indicated, or the spectrum of Candida is reportedly the fourth most common organism iso-
activity should be narrowed as much as possible. Cutaneous lated from the bloodstream.33 Although many human infec-
and corneal infections are usually treated with a combination of tions result from invasion of tissues by commensal yeasts in the
topical antiseptics or antifungal medications and systemic anti- face of immune suppression, horizontal transmission of Can-
fungal drugs until lesions resolve.8 dida and hospital-acquired infections have been described.34
Dogs and cats with Candida UTIs should initially be treated Whether Candida isolates from dogs or cats colonize humans
with antifungal drugs that achieve high concentrations in the is not known.
CHAPTER 67  Candidiasis 657

CASE EXAMPLE
Signalment: “Lizzie”, an 18-year-old female spayed domestic
shorthair from the San Francisco Bay area (Figure 67-3, A)
History: Lizzie was evaluated by the University of California,
Davis, Veterinary Medical Teaching Hospital for a 12-hour
history of dysuria and hematuria. The owner described
multiple episodes of urination and defecation outside the
litter box with stranguria, hematuria, and tenesmus. Lizzie was
also observed repeatedly grooming her perineum. The cat had
been diagnosed with diabetes mellitus 10 years previously, and
more recently with feline bronchial disease, a thoracolumbar
(T3 to L3) myelopathy, generalized osteoarthritis, chronic
constipation, bilateral cataracts, chronic kidney disease, and
recurrent bacterial UTIs (Streptococcus bovis group, Escherichia
coli, Enterococcus faecalis, then Aerococcus viridans). She also
had a cardiac murmur secondary to dynamic right ventricular A
outflow tract obstruction. The historical UTIs had been treated
with a variety of antimicrobial drugs selected based on the
results of aerobic bacterial urine culture and susceptibility
(most recently doxycycline, but also amoxicillin-clavulanic
acid or marbofloxacin in the past). Her current medications
consisted of prozinc insulin (3.5 units SC q12h), lactulose
(2 mL PO q12h), a soluble fiber supplement (1 teaspoon with
food twice daily), inhaled fluticasone (110-µg puff q12h),
theophylline (12 mg/kg PO q24h), and doxycycline (5 mg/
kg PO q12h). Her most recent blood work (collected 1 month
previously) showed a creatinine of 1.8 mg/dL (reference range,
1.1-2.2 mg/dL) and a BUN of 34 (reference range, 18-33 mg/
dL), which were similar to values obtained over the course
of the previous 5 years. She was an indoor-only cat and was
fed a commercial cat food with occasional turkey meat from
the delicatessen. Her appetite was good, there had been no
weight loss noted, her water consumption was unchanged,
and there had been no vomiting or diarrhea. She had not
been vaccinated for the past 4 years but was on regular flea
B
preventative medication (nitenpyram as well as fipronil).
Physical Examination: FIGURE 67-3  A, 18-year-old female spayed domestic shorthair with a Candida
Body Weight: 4.1 kg. albicans urinary tract infection, as well as diabetes mellitus, feline bronchial disease,
General: Quiet, alert and responsive, T = 100.5°F (38.1°C), a myelopathy, and generalized osteoarthritis. B, Bladder lumen as viewed by cystos-
copy. The urine contains large amounts of fluffy white debris that was loosely adherent
HR = 170 beats/min, RR = 28 breaths/min, CRT <1 s, pale pink
to the bladder wall.
mucous membranes.
Integument: A dull, slightly unkempt haircoat with mild scale
was noted. and irregular. The urinary bladder was approximately 5 cm
Eyes, Ears, Nose, and Throat: Immature cataracts were in diameter and soft. Severe erythema was noted around
present bilaterally, and all teeth were absent on examination the vulva.
of the oral cavity. No other clinically significant abnormalities Lymph Nodes: All were <1 cm in diameter.
were detected. Laboratory Findings: Urinalysis: SGr 1.030; pH 6.0, 75 mg/dL
Musculoskeletal: BCS was 4/9, and there was mild generalized protein, 1000 mg/dL glucose, no ketones or bilirubin, 250
muscle atrophy. The cat was reluctant to ambulate, and erythrocytes/µL hemoprotein, >100 WBC/HPF, >100 RBC/
appeared painful on manipulation of the elbows, stifles, HPF, few transitional and squamous epithelial cells, many
and coxofemoral joints bilaterally. Crepitus was detected on budding yeast organisms
manipulation of the tarsus. Microbiologic Testing: Aerobic bacterial urine culture: 105
Cardiovascular and Respiratory Systems: A grade II/VI left organisms/mL Candida albicans were cultured.
parasternal heart murmur was auscultated. Pulses were Yeast antifungal drug susceptibility panel: Susceptible to 5-
strong and synchronous. No other abnormalities were flucytosine (0.5 µg/mL), caspofungin (0.25 µg/mL),
detected. f­luconazole (0.25 µg/mL), itraconazole (0.06 µg/mL), vori-
Abdominal Palpation: The abdomen was soft and nonpainful. conazole (0.015 µg/mL). Minimum inhibitory concentrations
Both kidneys were small (approximately 2 cm in diameter) for amphotericin B and posaconazole were 0.5 µg/mL

Continued
658 SECTION 3  Fungal and Algal Diseases

and 0.03 µg/mL, respectively (no interpretative criteria irregular urinary bladder wall that contained hypoechoic,
available). ill-defined nodules, and thick sediment within the urinary
Diagnosis: Susceptible C. albicans UTI. bladder lumen. Cystoscopy revealed a large amount of
Treatment: Lizzie was treated with fluconazole (50 mg PO q12h). white, fluffy debris adherent to an edematous bladder wall
At a recheck examination 10 days later, the owner reported as well as in the lumen (Figure 67-3, B). A 10F Foley cath-
ongoing pollakiuria and incontinence. A fungal urine culture eter was placed over a 0.025-inch guidewire. The bladder
again revealed 105 C. albicans/mL with a similar susceptibility was flushed and the majority of the debris removed. A total
profile. Addition of flucytosine was recommended (250 mg of 47 mL of 1% clotrimazole solution was infused into the
PO q8h) but was considered unaffordable to the owners. bladder, and the solution was allowed to remain in place for
Treatment was continued for another month with little clinical 1 hour. The solution was removed and Lizzie was allowed
improvement and persistence of infection. A blood glucose to recover from anesthesia. Treatment with fluconazole was
curve revealed two-hourly blood glucose concentrations that continued, although the dose was lowered (25 mg PO q12h).
were “high”, 470, 368, 300, 383, and 423 mg/dL, respectively, At a recheck 2 weeks later, the owners reported that Lizzie
so the insulin dose was increased to 4 U q12h. Two weeks was more active and was no longer pollakiuric, although she
later the owner described some improvement in the urinary still urinated outside the litter box and intermittent vomiting
incontinence and reduction in stranguria, but again 105 had been noted. A CBC, biochemistry panel, and urinalysis
C. albicans/mL was cultured from the urine, and persistent revealed nonregenerative anemia (HCT 21.2%); lymphope-
urine scalding was noted around the perineum and on the nia (349 cells/µL); monocytopenia (70 cells/µL); increased
pelvic limbs on physical examination. Body weight was activities of serum ALT (1028 U/L; RR, 27-101 U/L), AST
3.8 kg, and a blood glucose curve revealed two-hourly blood (226 U/L; RR, 17-58 U/L), and ALP (223 U/L; RR, 14-17 U/L);
glucose concentrations that were 457, 301, 209, 227, 300, and hypercholesterolemia (324 U/L; RR, 89-258 U/L); and hyper-
296 mg/dL, respectively. bilirubinemia (0.7 mg/dL; RR, 0-0.2 mg/dL). Aerobic bacterial
Topical treatment with clotrimazole was elected. Before the urine culture revealed more than 105 CFU/mL of hemolytic
procedure was performed, a preanesthetic evaluation was E. coli, and fungal culture of the urine was negative. The fluco-
done. A CBC showed a normocytic, normochromic, non- nazole was discontinued, after which the vomiting ceased and
regenerative anemia (HCT 28.3%; RR, 30%-50%), mature liver enzyme activities returned to reference ranges. Multiple
neutrophilia (15,206 cells/µL; RR, 2000-9000 cells/µL), lym- bacterial UTIs were diagnosed over the following 6 months,
phopenia (919 cells/µL; 1000-7000 cells/µL), and monocy- but Candida was never again isolated from the urine.
topenia (33 cells/µL; 50-600 cells/µL). A serum biochemistry Comments: In this cat, the C. albicans UTI may have occurred
profile was unremarkable with the exceptions of a BUN con- secondary to a combination of factors that likely included
centration of 49 mg/dL (18-33 mg/dL) and a glucose concen- poorly controlled diabetes mellitus, chronic kidney disease,
tration of 314 mg/dL (63-118 mg/dL). Urinalysis showed a spe- neurologic disease, and repeated treatment with broad-
cific gravity of 1.022; pH 6.0, 25 mg/dL protein, 1000 mg/dL spectrum antimicrobial drugs. Systemic antifungal drug
glucose, 0-2 WBC/HPF, 0-2 RBC/HPF, and moderate numbers treatment and improved control of diabetes mellitus were
of budding yeast organisms and fungal structures. Thoracic initially used as treatment strategies but failed, probably
radiographs revealed mild cardiomegaly and a diffuse, static because of the large mats of fungus that were adherent to
bronchial pattern consistent with the previous diagnosis the bladder wall. Fortunately, a single topical clotrimazole
of feline bronchial disease. Abdominal ultrasound findings treatment resulted in resolution of the yeast infection. The
included an enlarged and hyperechoic liver, mild diffuse hepatopathy that developed was thought to be secondary
thickening of the gastrointestinal mucosa and submucosa, to fluconazole toxicity, since it resolved when the drug was
decreased renal corticomedullary distinction bilaterally with discontinued.
thickening of the renal cortices, a markedly thickened and

SUGGESTED READINGS 3. Heseltine JC, Panciera DL, Saunders GK. Systemic candidiasis in a
dog. J Am Vet Med Assoc. 2003;223:821-824:810.
Cheng SC, Joosten LA, Kullberg BJ, et al. Interplay between Candida 4. Ong RK, Raisis AL, Swindells KL. Candida albicans peritonitis in a
albicans and the mammalian innate host defense. Infect Immun. dog. J Vet Emerg Crit Care (San Antonio). 2010;20:143-147.
2012;80:1304-1313. 5. Rogers CL, Gibson C, Mitchell SL, et al. Disseminated candidiasis
Ong RK, Raisis AL, Swindells KL. Candida albicans peritonitis in a dog. secondary to fungal and bacterial peritonitis in a young dog. J Vet
J Vet Emerg Crit Care (San Antonio). 2010;20:143-147. Emerg Crit Care (San Antonio). 2009;19:193-198.
Pressler BM, Vaden SL, Lane IF, et al. Candida spp. urinary tract infec- 6. Jin Y, Lin D. Fungal urinary tract infections in the dog and cat:
tions in 13 dogs and seven cats: predisposing factors, treatment, and a retrospective study (2001-2004). J Am Anim Hosp Assoc.
outcome. J Am Anim Hosp Assoc. 2003;39:263-270. 2005;41:373-381.
7. Boutilier P, Carr A. Fungal colonization and failure of a long-term
REFERENCES gastrostomy tube in a cat. Can Vet J. 2005;46:709-710.
1. Barnett JA. A history of research on yeasts 12: medical yeasts part 8. Mueller RS, Bettenay SV, Shipstone M. Cutaneous candidiasis
1, Candida albicans. Yeast. 2008;25:385-417. in a dog caused by Candida guilliermondii. Vet Rec. 2002;150:
2. Pressler BM, Vaden SL, Lane IF, et al. Candida spp. urinary tract 728-730.
infections in 13 dogs and seven cats: predisposing factors, treat- 9. Dale JE. Canine dermatosis caused by Candida parapsilosis. Vet
ment, and outcome. J Am Anim Hosp Assoc. 2003;39:263-270. Med Small Anim Clin. 1972;67:548-549.
CHAPTER 67  Candidiasis 659

10. Ozawa H, Okabayashi K, Kano R, et  al. Rapid identification 23. Karkowska-Kuleta J, Rapala-Kozik M, Kozik A. Fungi pathogenic
of Candida tropicalis from canine cystitis. Mycopathologia. to humans: molecular bases of virulence of Candida albicans, Cryp-
2005;160:159-162. tococcus neoformans and Aspergillus fumigatus. Acta Biochim Pol.
11. Sykes JE. Unpublished observations, 2012. 2009;56:211-224.
12. Gerding Jr PA, Morton LD, Dye JA. Ocular and disseminated 24. Cheng SC, Joosten LA, Kullberg BJ, et  al. Interplay between
candidiasis in an immunosuppressed cat. J Am Vet Med Assoc. Candida albicans and the mammalian innate host defense. Infect
1994;204:1635-1638. Immun. 2012;80:1304-1313.
13. Mohri T, Takashima K, Yamane T, et  al. Purulent pericarditis 25. Holoymoen JI, Bjerkas I, Olberg IH, et  al. Disseminated can-
in a dog administered immune-suppressing drugs. J Vet Med Sci. didiasis (moniliasis) in a dog. A case report. Nord Vet Med.
2009;71:669-672. 1982;34:362-367.
14. Ochiai K, Valentine BA, Altschul M. Intestinal candidiasis in a dog. 26. Skoric M, Fictum P, Slana I, et  al. A case of systemic mycosis in
Vet Rec. 2000;146:228-229. a Hovawart dog due to Candida albicans. Veterinarni Medicina.
15. Rodriguez F, Fernandez A, Espinosa de los Monteros A, et  al. 2011;56:260-264.
Acute disseminated candidiasis in a puppy associated with parvovi- 27. Kuwamura M, Ide M, Yamate J, et  al. Systemic candidiasis in a
ral infection. Vet Rec. 1998;142:434-436. dog, developing spondylitis. J Vet Med Sci. 2006;68:1117-1119.
16. Langheinrich KA, Nielsen SW. Histopathology of feline panleuko- 28. Linek J. Mycotic endophthalmitis in a dog caused by Candida albi-
penia: a report of 65 cases. J Am Vet Med Assoc. 1971;158:suppl cans. Vet Ophthalmol. 2004;7:159-162.
2:863+. 29. Rosenvinge FS, Dzajic E, Knudsen E, et al. Performance of matrix-
17. Matsuda K, Sakaguchi K, Kobayashi S, et al. Systemic candidiasis assisted laser desorption-time of flight mass spectrometry for iden-
and mesenteric mast cell tumor with multiple metastases in a dog. tification of clinical yeast isolates. Mycoses. 2012.
J Vet Med Sci. 2009;71:229-232. 30. Toll J, Ashe CM, Trepanier LA. Intravesicular administration of
18. Miller WW, Albert RA. Ocular and systemic candidiasis in a cat. clotrimazole for treatment of candiduria in a cat with diabetes mellitus.
J Am Anim Hosp Assoc. 1988;24:521-524. J Am Vet Med Assoc. 2003;223:1156-1158:1129.
19. Lorenzini R, De Bernardis F. Antemortem diagnosis of an apparent 31. Forward ZA, Legendre AM, Khalsa HD. Use of intermittent blad-
case of feline candidiasis. Mycopathologia. 1986;93:13-14. der infusion with clotrimazole for treatment of candiduria in a dog.
20. Brown MR, Thompson CA, Mohamed FM. Systemic candidia- J Am Vet Med Assoc. 2002;220:1496-1498:1474-1495.
sis in an apparently immunocompetent dog. J Vet Diagn Invest. 32. Azie N, Neofytos D, Pfaller M, et al. The PATH (Prospective Anti-
2005;17:272-276. fungal Therapy) Alliance registry and invasive fungal infections: update
21. Ling GV, Norris CR, Franti CE, et al. Interrelations of organism 2012. Diagn Microbiol Infect Dis. 2012;73:293-300.
prevalence, specimen collection method, and host age, sex, and 33. Pappas PG. Invasive candidiasis. Infect Dis Clin North Am.
breed among 8,354 canine urinary tract infections (1969-1995). 2006;20:485-506.
J Vet Intern Med. 2001;15:341-347. 34. Ben Abdeljelil J, Saghrouni F, Emira N, et  al. Molecular typ-
22. Norris CR, Williams BJ, Ling GV, et al. Recurrent and persistent ing of Candida albicans isolates from patients and health care
urinary tract infections in dogs: 383 cases (1969-1995). J Am Anim workers in a neonatal intensive care unit. J Appl Microbiol.
Hosp Assoc. 2000;36:484-492. 2011;111:1235-1249.
CHAPTER 68

Miscellaneous Fungal Diseases


Amy M. Grooters

I
n contrast to the well-recognized fungal pathogens that
cause endemic mycoses (such as Blastomyces, Histo- Overview of Phaeohyphomycoses
plasma, and Cryptococcus), miscellaneous fungal patho- Causes: Alternaria, Bipolaris, Cladophialophora, Curvularia,
gens comprise large groups of fungi with unfamiliar names Exophiala, Fonsecaea, Moniliella, Phialophora, Ramichlo-
that traditionally have caused disease only sporadically. These ridium, Ulocladium, and Scolecobasidium, among many
opportunistic fungi have low inherent virulence and typically others
cause infection only when normal host resistance mechanisms
Affected Hosts: A variety of animal hosts that include dogs,
are compromised. Over the past 30 years, these opportunists
cats, horses, and humans
have become increasingly important in human patients because
immunocompromise associated with chemotherapy, organ Geographic Distribution: Worldwide
transplantation, and HIV infection has become more preva- Mode of Transmission: Cutaneous inoculation of organisms
lent. Similarly, over the past 5 to 10 years, the frequency with from the environment
which opportunistic fungal infections are encountered in small
Major Clinical Signs: Localized cutaneous nodules that may
animal patients has increased significantly in association with
appear pigmented, often on the nose or digits; occasion-
the use of multiagent immunosuppressive therapy (especially
ally meningoencephalitis, with associated neurologic
with cyclosporine) to treat immune-mediated disease in dogs.
signs
Thus, veterinarians are increasingly likely to encounter patients
infected by an opportunistic fungal pathogen with which they Differential Diagnosis: Neoplasia, cryptococcosis, blastomy-
are unfamiliar. cosis, sporotrichosis, hyalohyphomycosis, zygomycosis,
Unlike the common endemic fungal pathogens (for which mycobacterial infections, actinomycosis, nocardiosis
a definitive diagnosis can often be made simply by visualiz- Human Health Significance: Direct transmission from ani-
ing the unique morphologic features of the organism in cyto- mals to humans has not been described; humans develop
logic or histologic samples), opportunistic fungi can only be infections from environmental sources.
identified to genus and species level by culture or molecular
methods. However, they can be assigned to categories based
on their morphologic features in tissue, such as pigmentation,
hyphal diameter, and frequency of septation. These catego-
ries include phaeohyphomycosis (pigmented hyphal or yeast Phaeohyphomycosis
forms), hyalohyphomycosis (nonpigmented hyphal forms),
eumycotic mycetoma (fibrosing granuloma with black or Etiology and Epidemiology
white tissue grains consisting of aggregates of pigmented or
nonpigmented fungi, respectively), and zygomycosis (wide, The term “phaeohyphomycosis” refers to cutaneous, subcuta-
infrequently septate, nonpigmented hyphae associated with neous, cerebral, or disseminated infections caused by pigmented
pyogranulomatous and eosinophilic inflammation). Because (also known as dematiaceous) fungi that contain melanin in
of its similarities to pythiosis and lagenidiosis, zygomycosis is their cell walls.1 Infection usually results from cutaneous inoc-
discussed in Chapter 69. Although identification of a specific ulation. Fungal genera that have been identified as agents of
pathogen after culture is ideal, classification of opportunistic phaeohyphomycosis in veterinary patients include Alternaria,
mycoses based on the categories just listed is usually sufficient ­Bipolaris, Cladophialophora, Curvularia, Exophiala, Fonse-
to allow the clinician to make a reasonable prediction about caea, ­Moniliella, ­Phialophora, Ramichloridium, Ulocladium,
clinical course and prognosis and to make appropriate treat- and ­Scolecobasidium, among others (Table 68-1).
ment decisions. Because many opportunistic fungi are com-
mon contaminants and can normally be found on skin, nasal Clinical Features
mucosa, and other nonsterile sites, culture- or PCR-based
identification of a potential opportunistic fungal pathogen The most common clinical manifestations of phaeohyphomycosis
from a skin specimen, nasal swab, or exudate should not be in immunocompetent small animals are lesions associated with
considered evidence of fungal infection unless there is sup- the digits, pinnae, nasal planum, or nasal cavity in cats (Figure
portive histologic or cytologic evidence of tissue invasion by a 68-1). 2-8 Cutaneous lesions occur less often in dogs.9 In addition,
morphologically compatible organism. granulomatous meningoencephalitis caused by pigmented fungi

660
CHAPTER 68  Miscellaneous Fungal Diseases 661

TABLE 68-1
Causative Agents and Histologic/Cytologic Characteristics Associated with Miscellaneous Fungal and Pseudofungal Pathogens
Disease Causative Agents Histologic and Cytologic Characteristics
Phaeohyphomycosis Alternaria, Bipolaris, Phialophora, Clado- Pyogranulomatous inflammation associated with pigmented,
phialophora (Cladosporium), Curvu- irregularly septate hyphae or yeast-like cells that may be
laria, Exophiala, Fonsecaea, Moniliella, solitary or may cluster in small groups or chains
Ochroconis, Ramichloridium, others
Hyalohyphomycosis Acremonium, Fusarium, Geotrichum, Geo- Pyogranulomatous inflammation associated with hyphal ele-
smithia, Paecilomyces, Phialosimplex, ments that have nonpigmented (transparent, hyaline) walls;
Pseudallescheria, Sagenomella, Scedo- Phialosimplex may cause yeast-like forms in tissue
sporium, Schizophyllum, others
Mycetoma Curvularia Pyogranulomatous inflammation associated with aggregates
(black-grain) of pigmented fungal organisms (which appear grossly as
pigmented tissue grains)
Mycetoma Pseudallescheria boydii, Acremonium Pyogranulomatous inflammation associated with aggregates
(white-grain) of nonpigmented fungal organisms (which appear grossly
as nonpigmented tissue grains)
Pythiosis Pythium insidiosum Pyogranulomatous and eosinophilic inflammation associ-
ated with broad (2-7 µm), infrequently septate hyphae (see
Chapter 69)
Lagenidiosis Lagenidium Pyogranulomatous and eosinophilic inflammation associated
with broad (4-25 µm), infrequently septate hyphae (see
Chapter 69)
Zygomycosis Conidiobolus spp., Basidiobolus rana- Pyogranulomatous and eosinophilic inflammation associated
rum in dogs; Rhizomucor, Mucor, and with broad (5-20 µm), infrequently septate hyphae with
Cokeromyces in cats thick prominent eosinophilic sleeve (see Chapter 69)
Aspergillosis Aspergillus terreus, A. deflectus, A. flavi- Suppurative to granulomatous inflammation associated with
pes, A. fumigatus, others multiple, nonpigmented, 3- to 6-µm, septate hyphae with
parallel walls and 45-degree angle branching (see Chapter 65)
Candidiasis Candida albicans, other Candida spp. Suppurative inflammation with numerous 2- to 6-µm oval
yeasts, pseudohyphae (chains of oval yeast cells), and true
hyphae (see Chapter 67)

(especially Cladophialophora bantiana) have been described


both in dogs and cats.10-14 In immunocompromised patients, the
most common presentation appears to be multifocal cutaneous
lesions in dogs treated with multiagent immunosuppressive ther-
apy, especially that which includes cyclosporine.15-17
Animals with phaeohyphomycosis typically have cutaneous
nodules or a visible nasal mass. Infected tissues may appear grossly
pigmented, and thus masses may be confused with melanomas.
Phaeohyphomycoses tend to be locally invasive and may extend to
involve regional lymph nodes (Figure 68-2). Systemic dissemina-
tion more often occurs in animals treated with immunosuppres-
sive drugs, but disseminated infections have also been described in
apparently immunocompetent patients.18,19 Animals with central
nervous system (CNS) phaeohyphomycosis may have neurologic
signs such as obtundation, seizures, abnormal placing reactions,
circling, tremors, ataxia, and abnormal cranial nerve function.

Diagnosis
Laboratory and Imaging Abnormalities
FIGURE 68-1  Left distal thoracic limb of an 8-year-old male neutered Burmese cat Results of a CBC, biochemistry panel, and urinalysis in animals
with digital phaeohyphomycosis. (Courtesy University of California, Davis, Veterinary Der- with cutaneous phaeohyphomycoses are typically unremark-
matology Service.) able. Animals with disseminated disease can have abnormalities
662 SECTION 3  Fungal and Algal Diseases

A B

FIGURE 68-2  Popliteal lymphadenopathy and abscessation caused by Curvularia


spp. infection in a 1-year-old female Yorkshire terrier with cutaneous phaeohyphomycosis.
(Courtesy Amy Grooters, Louisiana State University, Baton Rouge, LA.) C

a­ ssociated with underlying immunosuppressive illness or drug


treatment. Imaging with computed tomography (CT) or MRI in
animals with CNS involvement may reveal focal or multifocal
contrast-­enhancing lesions that may be associated with hydro-
cephalus (Figure 68-3).

Cytologic and Histologic Findings


On histologic or cytologic examination, fungi that cause pha-
eohyphomycosis appear as dark-walled, irregularly septate
hyphae or as yeast-like cells, solitary or in small groups or
chains (­Figure 68-4). Pigmentation may not always be appar-
ent on cytologic examination (Figure 68-5). The presence of
melanin in the walls of lightly-pigmented hyphae can be con-
firmed by the examination of unstained sections, by lowering
the microscope condenser during examination, or by utilization
of a Fontana-Masson stain for melanin.
D
Culture
FIGURE 68-3  Axial T1 postcontrast, T2, and FLAIR magnetic resonance images (A,
Organisms that cause phaeohyphomycosis are readily isolated B, and C, respectively) of the brain of a 6-year-old male neutered Border collie mix that
on routine fungal culture of infected tissue biopsies as well as was evaluated for neurologic signs including seizures and progressive obtundation. In the
from fine-needle aspirate specimens from infected lymph nodes. images shown, there is bilateral but asymmetric ventricular dilation that is severe on the
Further identification is based on colony and conidial mor- right and mild to moderate on the left (asymmetric hydrocephalus). Multifocal regions
phology (see also Figure 4-7, B) and PCR and sequencing of of contrast enhancement are also present (A). Some of these regions appeared nodular;
ribosomal RNA genes. Because pigmented fungi are common others had a ring-like appearance. There is also marked contrast enhancement of the ven-
laboratory contaminants and can sometimes be isolated from tricular ependymal lining. The regions of contrast enhancement correlate with regions of
nonsterile sites on healthy animals (such as skin), positive cul- T2 and FLAIR hyperintensity (B and C) and were isointense on precontrast T1-weighted
tures from potentially contaminated sites should only be con- sequences (not shown). Meningoencephalitis caused by Cladophialophora bantiana and
secondary hydrocephalus was diagnosed on brain biopsy and at necropsy (D). Olive-green
sidered significant if accompanied by cytologic or histologic
pigmented masses can be seen on both the left and right sides of the brain. Interestingly,
evidence of fungal infection with a morphologically compatible the dog also had cutaneous protothecosis. (Courtesy University of California, Davis, Veteri-
organism. nary Neurology and Anatomic Pathology Services.)

Treatment and Prognosis


3 to 6 months after surgery, because recurrence of disease at the
Pigmented fungi are often poorly responsive to medical therapy, surgical site is common.
in part because melanin is a virulence factor. Aggressive surgical For nonresectable lesions, treatment with itraconazole
resection is the treatment of choice for solitary cutaneous pha- (10 mg/kg/day) often results in partial or complete resolution of
eohyphomycosis lesions. The surgeon should attempt to obtain cutaneous lesions, but recurrence is very common, so prolonged
wide margins at the time of the initial surgery. Digit amputation courses (6 to 12 months) should be recommended. Voricon-
is usually indicated for lesions of the distal phalanx. Medical azole or posaconazole may be more effective than itraconazole
therapy with itraconazole or posaconazole is recommended for for the treatment of phaeohyphomycosis but are significantly
CHAPTER 68  Miscellaneous Fungal Diseases 663

Public Health Aspects


Phaeohyphomycosis in humans clinically resembles that in dogs
and cats and may include localized cutaneous lesions in immu-
nocompetent hosts; life-threatening CNS infections in immuno-
compromised or apparently immunocompetent hosts (especially
brain abscesses caused by C. bantiana); and, rarely, dissemi-
nated phaeohyphomycosis, primarily in immunocompromised
hosts.21 Infections caused by pigmented fungi are acquired from
the environment. Although there is no evidence of transmission
between mammalian hosts, routine precautions (such as wear-
ing gloves when handling infected tissues or exudate) should be
followed. Precautions should also be taken to prevent needle-
stick injuries if phaeohyphomycosis is suspected.
FIGURE 68-4  Histopathology showing small groups of pigmented, yeast-like fungal
organisms in tissue from a nasal mass in a cat with phaeohyphomycosis; H&E stain. (Cour-
tesy Amy Grooters, Louisiana State University, Baton Rouge, LA.) Overview of Hyalohyphomycoses
Causes: Acremonium, Fusarium, Geotrichum, Paecilomyces,
Pseudallescheria, Sagenomella, Scedosporium, others
Affected Hosts: A variety of animal hosts that include dogs,
cats, and humans
Geographic Distribution: Worldwide
Mode of Transmission: Inhalation or cutaneous inoculation of
organisms in the environment
Major Clinical Signs: Keratitis; cutaneous lesions; osteomyeli-
tis; pneumonia; disseminated disease with involvement of
lymph nodes, kidneys, liver, spleen, bone, intervertebral
discs, and/or CNS
Differential Diagnosis: Neoplasia, aspergillosis, penicilliosis,
phaeohyphomycoses, sporotrichosis, cryptococcosis,
blastomycosis, candidiasis, endemic mycoses, nocardio-
sis, actinomycosis, mycobacteriosis, zygomycosis
Human Health Significance: Direct transmission from ani-
mals to humans has not been described; humans develop
FIGURE 68-5  Impression smear from the brain of the dog in Figure 68-3. Septate, infections from environmental sources.
branching hyphae are present, but pigmentation is not clearly apparent. Wright’s stain,
1000× magnification. (Courtesy University of California, Davis, Veterinary Pathology
Service.)
Hyalohyphomycosis

Etiology and Epidemiology


more expensive. In addition, voriconazole administration is
not recommended in cats because of significant adverse effects The term “hyalohyphomycosis” refers to infections caused by
(see Chapter 9). Recently, long-term voriconazole therapy fungi that are nonpigmented (hyaline or transparent) in tissue.
(7 to 10 mg/kg/day for 10 to 12 months) was used success- Genera that have been described as agents of hyalohyphomyco-
fully to treat intracranial phaeohyphomycosis in one dog10 and sis in veterinary patients include Fusarium, Acremonium, Pae-
mycotic peritonitis caused by Exophiala in another.20 Early cilomyces (especially Paecilomyces variotii and ­Paecilomyces
combination therapy with amphotericin B, flucytosine, and lilacinus), Pseudallescheria boydii (anamorph Scedosporium
extended-spectrum azoles has been recommended for treatment apiospermum), Sagenomella, Phialosimplex, Geosmithia, and
of human patients with CNS infections,21 but flucytosine can be Geomyces, among others (see Table 68-1). By convention,
prohibitively expensive and is generally not tolerated by dogs infections caused by Aspergillus and Penicillium species are not
(see Chapter 9). included in the term “hyalohyphomycosis,” because aspergil-
In the author’s experience, cutaneous phaeohyphomycosis losis and penicilliosis can usually be identified as such based on
that occurs in dogs receiving immunosuppressive drug therapy their clinicopathologic features. In general, hyalohyphomycosis
can be resolved in many cases with itraconazole (10 mg/kg/day) occurs more often in dogs than in cats.
administered for at least 6 months if the immunosuppressive
drug therapy can be tapered quickly. However, dissemination of Clinical Features
disease can also occur, sometimes despite appropriate antifun-
gal therapy. Discontinuation of cyclosporine treatment in these Hyalohyphomycosis ranges from a local disease confined to the
animals appears to be important for achieving a good outcome. skin (Figure 68-6), nasal mucosa, or cornea to osteomyelitis,
664 SECTION 3  Fungal and Algal Diseases

FIGURE 68-7  Fine needle aspirate cytology from a lytic bone lesion in a 4-year-old
female spayed mixed breed dog with hyalohyphomycosis. Note the septate, nonpig-
mented fungal hyphae present within the macrophage; Wright-Giemsa stain, bar =10 µm.
(Courtesy Kaikhushroo Banajee, Louisiana State University, Baton Rouge, LA.)

FIGURE 68-6  Scedosporium prolificans infection in a domestic shorthair cat. There are
multiple draining skin lesions and soft tissue swelling. (Courtesy Spencer Jang, University
of California, Davis.)

pneumonia, and disseminated disease that can involve one or


more organs including the kidneys, bone marrow, lymph nodes,
liver, spleen, bones, intervertebral discs, and CNS, sometimes
with associated fungemia.22-30
Traditionally, the disseminated and corneal forms of hyalo-
hyphomycosis have been most commonly identified. Therefore,
animals with no overt signs of systemic disease that present with
cutaneous or bone lesions that contain nonpigmented hyphae
should still be evaluated for occult lesions in the chest and
abdomen. Like phaeohyphomycosis, hyalohyphomycosis is an FIGURE 68-8  Cytologic appearance of Paecilomyces variotii growing in culture in the
increasingly common cause of multifocal skin lesions in dogs laboratory. Lactophenol cotton blue, 1000× magnification. (Courtesy Spencer Jang, Uni-
receiving multiagent immunosuppressive therapy. These dogs versity of California, Davis.)
may or may not have accompanying extracutaneous lesions.
University of Texas Health Science Center at San Antonio) and
Diagnosis may be useful for selection of treatments for difficult cases. Nev-
ertheless, evidence that supports a correlation between in vitro
Cytologic and Histologic Findings susceptibility data and clinical response to treatment is lacking.
Cytologically and histologically, fungi that cause hyalohypho-
mycosis are nonpigmented, frequently septate, branching hyphae Antigen Testing
that are often pleomorphic (Figures 68-7 and Figure 4-6). Special The Platelia Aspergillus galactomannan antigen ELISA assay is
stains such as periodic acid–Schiff and Gomori’s methenamine positive in some, but not all, animals with hyalohyphomyco-
silver may be used to identify organisms more readily. ses such as paecilomycosis. This results from antigenic cross-
reactivity (see Chapter 65).31 In most dogs with disseminated
Culture paecilomycosis that test positive, the magnitude of positive
The fungi that cause hyalohyphomycosis are readily isolated by results is somewhat lower than those in dogs with dissemi-
routine fungal culture methods from infected tissues as well as nated aspergillosis, but occasionally results are strongly posi-
from fine-needle aspirate specimens from infected lymph nodes, tive (galactomannan indices >6).
bones, or abdominal organs. Because these fungi are common
laboratory contaminants and can sometimes be isolated from Treatment and Prognosis
the skin or hair of healthy animals, positive cultures from non-
sterile sites should be considered significant only if accompa- Treatment of hyalohyphomycosis has traditionally been chal-
nied by cytologic or histologic evidence of fungal infection with lenging because many animals have disseminated disease. Drugs
a morphologically compatible organism. Species identification used most often to treat hyalohyphomycosis in small animals
is based on conidial morphology (Figure 68-8) and may require include itraconazole and amphotericin B, but different fungal
PCR and sequencing of ribosomal RNA genes. Species identi- species vary in their susceptibility to antifungal drugs, and some
fication may assist in selection of antifungal drug treatments, species, such as Pseudallescheria boydii and Paecilomyces lilaci-
because some species are predictably less susceptible to conven- nus, are relatively resistant to conventional antifungal drugs
tional antifungal drugs (see Treatment and Prognosis). Proper in  vitro.32,33 The newer triazoles, voriconazole and posacon-
identification and antifungal drug susceptibility testing are best azole, and the echinocandins, such as caspofungin, may be more
performed by laboratories that specialize in mycoses (e.g., the active against these fungi than itraconazole but are significantly
CHAPTER 68  Miscellaneous Fungal Diseases 665

more expensive. In contrast, Paecilomyces variotii is usually sus- bone that are characterized by the presence of colonies or aggre-
ceptible to amphotericin B but is less susceptible to voriconazole. gates of organisms that form “grains” in tissue. Actinomycotic
Although treatment of disseminated hyalohyphomycosis with mycetomas are caused by bacteria such as Actinomyces spp. or
antifungal drugs can prolong survival, clinical signs often recur Nocardia spp. (see Chapters 42 and 43), dermatophytic myceto-
even if signs initially resolve. Therefore, disseminated hyalohy- mas are caused by dermatophytes (see Chapter 58), and eumy-
phomycosis generally carries a guarded to poor prognosis. cotic mycetomas are caused by nondermatophyte fungi. Lesions
Cutaneous hyalohyphomycosis that develops in an animal result from traumatic implantation of soil organisms into tissue.
receiving immunosuppressive drug therapy may respond well to The grains or granules of eumycotic mycetomas are pigmented
oral azole antifungal drugs or may rapidly disseminate. Therefore, (for black-grain mycetoma, caused by dematiaceous fungi) or
fungal skin lesions that develop in immunocompromised patients hyaline (for white-grain mycetoma, caused by nonpigmented
should be treated aggressively, and a guarded prognosis should be fungi), depending on the type of fungal pathogen involved (see
offered. This author most often uses itraconazole (10 mg/kg/day) Table 68-1).
administered orally for at least 6 months. Other options include Black-grain mycetomas are most often caused by Curvu-
amphotericin B, voriconazole, or posaconazole. Combination laria species and typically are associated with chronic nonheal-
therapies have been recommended for difficult cases in human ing wounds and cutaneous nodules on the extremities. Lesions
patients.21 Discontinuation of cyclosporine and rapid tapering of often develop weeks to months after a traumatic incident in the
other immunosuppressive medications in these patients appears same area. Draining tracts are often present, and black grains
to be important for achievement of a good outcome. may be observed in the exudate. White-grain mycetomas, usu-
ally caused by Pseudallescheria boydii or Acremonium spp.,
Public Health Aspects most often occur as body-wall and/or intra-abdominal granulo-
mas that develop subsequent to surgical wound contamination
Hyalohyphomycosis in humans is acquired as a result of infection or dehiscence. Interestingly, the lesions associated with white-
by fungi in the environment. Keratitis and nailbed infections (ony- grain mycetoma may not be evident until months or even a year
chomycosis) can occur in immunocompetent humans. Invasive or more after the surgical event. Affected dogs may be evalu-
infections occur in immunocompromised patients such as trans- ated for a draining mass on the body wall or clinical signs of
plant recipients, patients with AIDS, and leukemic or neutropenic peritonitis.
patients. Although there is no evidence to suggest that transmission The treatment of choice for eumycotic mycetoma is aggres-
between mammalian hosts is possible, routine precautions (such as sive surgical excision of infected tissues, including amputation
wearing gloves when handling infected tissues or exudate) should if clinically indicated. Response to medical therapy is gener-
be followed. Precautions should especially be taken to prevent nee- ally poor, but administration of itraconazole, voriconazole, or
dle-stick injuries if hyalohyphomycosis is suspected. posaconazole may be considered as adjunctive therapy follow-
ing surgical excision when wide margins cannot be achieved.
Eumycotic Mycetoma Dissemination of eumycotic mycetoma beyond local tissues
is rare, but disease within the abdomen or other local tissues
The term “mycetoma” refers to localized mycotic or actino- may be extensive. The prognosis is guarded if complete surgical
mycotic infections of the skin, subcutaneous tissue, muscle, or resection is not possible.

CASE EXAMPLE Dermatologic Examination: Pitting edema and multifocal


ulcerative lesions were present on all four distal
extremities, with sloughing of multiple footpads (Figure
Signalment: “CJ”, a 1-year-old male intact boxer dog from 68-9, A and B). Some of the pad lesions were proliferative
central Louisiana and extended into the interdigital area (Figure 68-9, C).
History: CJ was initially evaluated for acute ataxia and abnormal Multiple light brown colored areas could be visualized
limb placement. Based on results of CSF analysis as well as MRI within the fibrinonecrotic material that covered the area
of the brain and spinal cord, inflammatory central nervous of the left metacarpal pad (see Figure 68-9, B). In addition,
system disease was diagnosed, and treatment with prednisone a 1-cm ulcerated lesion was present on the left pinna, a
(1 mg/kg PO q12h) was initiated. Despite a good initial response, crusted lesion was present on the tip of the tail, and a small
neurologic signs recurred 2 weeks later, at which time cytosine subcutaneous mass was present in the right saphenous
arabinoside (50 mg/m2 SC q12h, for four doses, repeated every area. Larger (3 cm) ulcerated lesions partially covered with
3 weeks) and azathioprine (1.6 mg/kg PO q48h) were added eschar were present on the medial aspect of the right
to the treatment protocol. Neurologic signs again improved, tarsus and the dorsal aspect of the left antebrachium. Major
but 7 weeks later, the dog was evaluated for ulcerative lesions differential diagnoses considered were opportunistic
on the distal extremities that had been present for 1 week and fungal infection and drug eruption.
lethargy that had been present for 3 days. Cytology and Histology Findings: Cytologic evaluation of
Physical Examination: the left pinnal lesion, subcutaneous right saphenous mass,
Body Weight: 29 kg. and right popliteal lymph node revealed septic neutrophilic
General: Alert and responsive, but painful on ambulation to pyogranulomatous inflammation with septate,
because of ulcerative lesions on all four feet. Vital parameters branching, 3- to 7-µm fungal hyphae, both extracellularly
were within normal limits. Peripheral lymph nodes were and within neutrophils. Pigmentation of hyphae was not
palpably normal. observed.

Continued
666 SECTION 3  Fungal and Algal Diseases

Histologic evaluation of punch biopsies obtained from cutane-


ous lesions on the left front foot, right hind foot, and right
forelimb revealed severe pyogranulomatous dermatitis with
intralesional septate, pigmented hyphae.
Diagnosis: Disseminated cutaneous phaeohyphomycosis
extending to regional lymphadenitis.
Treatment and Outcome: Pentoxifylline (30 mg/kg PO
q24h) was administered because of suspected vasculitis
associated with the cutaneous lesions. Antifungal therapy
with itraconazole (10 mg/kg PO q24h) was initiated, but
lesion progression was observed over the next several days,
so lipid-complexed amphotericin B treatment (2 mg/kg
diluted in 5% dextrose, IV, over several hours three times
weekly) was initiated. Unfortunately, 2 days later lesions had
progressed rapidly with appearance of new draining tracts
and abscess formation. Because of the poor prognosis and
A B pain associated with the lesions, the dog was euthanized.
Comments: As was noted here, pigmentation of hyphae may
not always be visible on cytologic examination of lesions
caused by phaeohyphomycosis, in part because hyphae and
the cells that phagocytize them often take up enough color
with the Wright-Giemsa stain to mask the pigmentation.
Therefore, in this case, evaluation of histologic samples
was important for differentiating phaeohyphomycosis
from hyalohyphomycosis. Culture would have been ideal
for definitive identification of the opportunistic fungal
organism that caused the infection in this dog, but was not
performed.

C
FIGURE 68-9  A-C, Phaeohyphomycosis of the left thoracic limb that developed
after multiagent immunosuppressive therapy in 1-year-old boxer with inflammatory
central nervous system disease. (Courtesy Amy Grooters, Louisiana State University, Baton
Rouge, LA.)

SUGGESTED READINGS 5. Beccati M, Vercelli A, Peano A, et  al. Phaeohyphomycosis by


Phialophora verrucosa: first European case in a cat. Vet Rec.
Grooters AM, Foil CS. Miscellaneous fungal infections. In: Greene CE, 2005;157:93-94.
ed. Infectious Diseases of the Dog and Cat. 4th ed. Philadelphia, PA: 6. Fondati A, Gallo MG, Romano E, et al. A case of feline phaeohypho-
WB Saunders; 2012:675-688. mycosis due to Fonsecaea pedrosoi. Vet Dermatol. 2001;12:297-301.
Naggie S, Perfect JR. Molds: hyalohyphomycosis, phaeohyphomycosis 7. Abramo F, Bastelli F, Nardoni S, et al. Feline cutaneous phaeohy-
and zygomycosis. Clin Chest Med. 2009;30:337-353. phomycosis due to Cladophialophora bantiana. J Feline Med Surg.
2002;4:157-163.
REFERENCES 8. Tennant K, Patterson-Kane J, Boag AK, et al. Nasal mycosis in two
1. Revankar SG, Sutton DA. Melanized fungi in human disease. Clin cats caused by Alternaria species. Vet Rec. 2004;155:368-370.
Microbiol Rev. 2010;23:884-928. 9. Rajeev S, Clifton G, Watson C, et al. Fonsecaea pedrosoi skin infec-
2. Dye C, Johnson EM, Gruffydd-Jones TJ. Alternaria species infec- tion in a dog. J Vet Diagn Invest. 2008;20:379-381.
tion in nine domestic cats. J Feline Med Surg. 2009;11:332-336. 10. Bentley RT, Faissler D, Sutherland-Smith J. Successful management
3. Maeda H, Shibuya H, Yamaguchi Y, et  al. Feline digital pha- of an intracranial phaeohyphomycotic fungal granuloma in a dog.
eohyphomycosis due to Exophiala jeanselmei. J Vet Med Sci. J Am Vet Med Assoc. 2011;239:480-485.
2008;70:1395-1397. 11. Giri DK, Sims WP, Sura R, et  al. Cerebral and renal phaeohy-
4. Knights CB, Lee K, Rycroft AN, et al. Phaeohyphomycosis caused phomycosis in a dog infected with Bipolaris species. Vet Pathol.
by Ulocladium species in a cat. Vet Rec. 2008;162:415-416. 2011;48:754-757.
CHAPTER 68  Miscellaneous Fungal Diseases 667

12. Anor S, Sturges BK, Lafranco L, et al. Systemic phaeohyphomyco- 23. Holahan ML, Loft KE, Swenson CL, et  al. Generalized calcino-
sis (Cladophialophora bantiana) in a dog—clinical diagnosis with sis cutis associated with disseminated paecilomycosis in a dog. Vet
stereotactic computed tomographic–guided brain biopsy. J Vet Dermatol. 2008;19:368-372.
Intern Med. 2001;15:257-261. 24. Caro-Vadillo A, Garcia-Real I, Paya-Vicens MJ, et al. Fungal rhini-
13. Bouljihad M, Lindeman CJ, Hayden DW. Pyogranuloma- tis caused by Scedosporium apiospermum in a Labrador retriever.
tous meningoencephalitis associated with dematiaceous fungal Vet Rec. 2005;157:175-177.
(­Cladophialophora bantiana) infection in a domestic cat. J Vet 25. Erne JB, Walker MC, Strik N, et al. Systemic infection with Geo-
Diagn Invest. 2002;14:70-72. myces organisms in a dog with lytic bone lesions. J Am Vet Med
14. Simmons JK, McManamon R, Rech RR, et al. ­Pathology in ­Practice. Assoc. 2007;230:537-540.
Necrotizing pyogranulomatous ­meningoencephalitis with 26. Tanaka H, Takizawa K, Baba O, et  al. Basidiomycosis: Schizo-
­intralesional fungal hyphae, consistent with ­Cladophialophora phyllum commune osteomyelitis in a dog. J Vet Med Sci.
bantiana. J Am Vet Med Assoc. 2010;236(3):295-297. 2008;70:1257-1259.
15. Dedola C, Stuart AP, Ridyard AE, et  al. Cutaneous Alternaria 27. Grant DC, Sutton DA, Sandberg CA, et al. Disseminated Geosmi-
infectoria infection in a dog in association with therapeutic immu- thia argillacea infection in a German shepherd dog. Med Mycol.
nosuppression for the management of immune-mediated haemo- 2009;47:221-226.
lytic anaemia. Vet Dermatol. 2010;21:626-634. 28. Armstrong PF, Sigler L, Sutton DA, et al. Fungal myelitis caused by
16. Swift IM, Griffin A, Shipstone MA. Successful treatment of dis- Phialosimplex caninus in an immunosuppressed dog. Med Mycol.
seminated cutaneous phaeohyphomycosis in a dog. Aust Vet J. 2012;50:509-512.
2006;84:431-435. 29. Marlar AB, Miller PE, Canton DD, et al. Canine keratomycosis: a
17. Herraez P, Rees C, Dunstan R. Invasive phaeohyphomycosis caused report of eight cases and literature review. J Am Anim Hosp Assoc.
by Curvularia species in a dog. Vet Pathol. 2001;38:456-459. 1994;30:331-340.
18. Coldrick O, Brannon CL, Kydd DM, et  al. Fungal pyelone- 30. Rampazzo A, Kuhnert P, Howard J, et  al. Hormographiella
phritis due to Cladophialophora bantiana in a cat. Vet Rec. aspergillata keratomycosis in a dog. Vet Ophthalmol. 2009;12:
2007;161:724-728. 43-47.
19. Elies L, Balandraud V, Boulouha L, et al. Fatal systemic phaeohy- 31. Garcia RS, Wheat LJ, Cook AK, et al. Sensitivity and specificity
phomycosis in a cat due to Cladophialophora bantiana. J Vet Med of a blood and urine galactomannan antigen assay for diagno-
A Physiol Pathol Clin Med. 2003;50(10):50-53. sis of systemic aspergillosis in dogs. J Vet Intern Med. 2012;26:
20. Murphy KF, Malik R, Barnes A, et al. Successful treatment of intra- 911-919.
abdominal Exophiala dermatitidis infection in a dog. Vet Rec. 32. Lackner M, de Hoog GS, Verweij PE, et al. Species-specific antifun-
2011;168:217. gal susceptibility patterns of Scedosporium and Pseudallescheria
21. Naggie S, Perfect JR. Molds: hyalohyphomycosis, phaeohyphomy- species. Antimicrob Agents Chemother. 2012;56(5):2635-2642.
cosis and zygomycosis. Clin Chest Med. 2009;30:337-353. 33. Pastor FJ, Guarro J. Clinical manifestations, treatment and out-
22. Foley JE, Norris CR, Jang SS. Paecilomycosis in dogs and horses come of Paecilomyces lilacinus infections. Clin Microbiol Infect.
and a review of the literature. J Vet Intern Med. 2002;16:238-243. 2006;12(10):948-960.
CHAPTER 69

Pythiosis, Lagenidiosis, and Zygomycosis


Amy M. Grooters

P
ythiosis, lagenidiosis, and zygomycosis are often grouped mammalian pathogens since 1999. In comparison to pythiosis
together because of similarities in their clinical presenta- and lagenidiosis, infections caused by the zygomycetes are rare.
tions and histologic characteristics (all three cause pyogran-
ulomatous and eosinophilic inflammation associated with large, PYTHIOSIS
infrequently septate hyphae with nonparallel walls). Despite their
clinicopathologic similarities, however, the pathogens that cause Etiology and Epidemiology
these infections are taxonomically diverse. Pythium insidiosum
and Lagenidium species are water molds in the class Oomycetes Pythium insidiosum is an aquatic oomycete that causes severe,
and as such are more closely related to red algae and Prototheca progressive gastrointestinal or cutaneous disease that is often
spp. than to true fungi, including the zygomycetes.1 Important fatal. The infective form of P. insidiosum is thought to be the
traits that distinguish oomycetes from fungi include the produc- motile flagellate zoospore, which is an asexual reproductive
tion of motile, flagellate zoospores that act as infective elements structure produced in wet environments in association with
in wet environments, and the fact that the oomycete cell mem- plant material.4 Zoospores of P. insidiosum are attracted to
brane generally lacks ergosterol.2 In addition, there are clini- damaged tissue, and likely cause infection by encystation on
cally relevant differences in prognosis, recommended treatment, and invasion of damaged skin or gastrointestinal mucosa.5 In
and epidemiology that make it important to distinguish among support of this mechanism of infection, clinical reports have
pythiosis, lagenidiosis, and zygomycosis. Although P. insidiosum long suggested an association between pythiosis and frequent
has been recognized as a pathogen in dogs and horses for more exposure to warm freshwater habitats. However, some infec-
than 25 years, Lagenidium species have only been recognized as tions occur in dogs with no history of access to lakes or ponds.
Globally, pythiosis occurs in tropical and subtropical cli-
mates, including Southeast Asia, eastern coastal Australia, and
South America (Figure 69-1). In the United States, the disease
Overview of Pythiosis
occurs most often in the Gulf Coast states, but it has also been
First Described: 1884, India (Smith)3, in horses
recognized throughout the South; along the East Coast as far
Cause: Pythium insidiosum (kingdom Stramenopila, class north as Maryland and New Jersey; in the Midwest including
Oomycota), related to algae and Prototheca spp. Missouri, Kansas, southern Illinois, and Indiana; and in the
Affected Hosts: Horses and dogs; less commonly cats, sheep, West in Arizona and California.6
cattle, exotic species, and humans Pythiosis is most common in young, large-breed dogs, espe-
cially Labrador retrievers and other outdoor working breeds.
Geographic Distribution: Tropical, subtropical, and some Infected dogs are typically evaluated by veterinarians in the
temperate regions worldwide, especially the Gulf Coast fall, winter, and early spring.7 Pythiosis is uncommon in cats
region of the United States compared to dogs, and specific breed and sex predilections have
Mode of Transmission: Likely penetration of damaged skin not been observed in the cases described to date. However,
or mucosa by motile zoospores, usually after exposure to the development of cutaneous pythiosis in very young animals
standing freshwater sources (less than 1 year of age) appears to occur more often in cats
Major Clinical Signs: Ulcerative, nodular, and mass-like cuta- than in dogs. For both species, affected animals are typically
neous lesions with draining tracts (cutaneous form); immunocompetent.
weight loss, anorexia, vomiting, diarrhea, hematochezia
(gastrointestinal form) Clinical Features
Differential Diagnosis: For suspected gastrointestinal pythio- The clinical signs associated with pythiosis are caused by gastro-
sis, differential diagnoses include neoplasia, zygomycosis, intestinal or cutaneous lesions, or by extension of disease into
histoplasmosis, eosinophilic gastroenteritis, or chronic regional lymph nodes or adjacent tissues. Generally, gastrointes-
intestinal foreign bodies; for suspected cutaneous pythio- tinal and cutaneous lesions do not occur together in the same ani-
sis, they include lagenidiosis, zygomycosis, mycobacterial mal. Disseminated pythiosis has only been described in one dog.8
infections, actinomycosis, or furunculosis.
Human Health Significance: P. insidiosum is a rare cause of Gastrointestinal Pythiosis
disease in human patients; infection is not transmitted Gastrointestinal pythiosis in dogs is characterized by severe,
directly from affected animals to people. segmental, transmural thickening of the stomach wall (Fig-
ure 69-2), small intestine, colon, rectum, or, rarely, the

668
CHAPTER 69  Pythiosis, Lagenidiosis, and Zygomycosis 669

FIGURE 69-1  Worldwide geographic distribution of pythiosis.

abdomen. Invasion of mesenteric vessels may result in bowel


ischemia, infarction, perforation, or acute hemoabdomen. In
addition, infection may extend into adjacent tissues such as
the pancreas or the uterus. Gastrointestinal pythiosis is rare
in cats but has been described in two young adult male cats
with focal intestinal lesions that were amenable to surgical
resection.11
Dogs with gastrointestinal pythiosis typically have a history
of chronic progressive weight loss, vomiting, diarrhea, anorexia,
and sometimes hematochezia, depending on the location of the
lesions. Regurgitation may be present in dogs with esophageal
involvement. Physical examination usually reveals a poor body
condition and sometimes a palpable abdominal mass. Signs of
systemic illness such as lethargy or depression are absent unless
intestinal obstruction, infarction, or perforation occurs.

Cutaneous Pythiosis
Cutaneous pythiosis in dogs occurs most often at the base of the
tail, or on the extremities, ventral neck, or perineum,8,12 and is
FIGURE 69-2  Gastric pythiosis in a 1-year-old male neutered mixed breed dog. Note characterized by nonhealing wounds and invasive masses that
extreme thickening of the gastric wall. (Courtesy Amy Grooters, Louisiana State University, contain ulcerated nodules and draining tracts (Figure 69-3). In
Baton Rouge, LA.) contrast to gastrointestinal pythiosis, regional lymphadenopa-
thy associated with cutaneous pythiosis often reflects extension
of infection rather than just reactive hyperplasia. Extension of
esophagus, and it is not uncommon to find multiple segmental cutaneous disease to tissues other than regional lymph nodes
lesions in the same patient.7,9,10 The most commonly affected is rare, but the author has observed a single, focal pulmonary
regions of the gastrointestinal tract are the gastric outflow lesion caused by P. insidiosum in one dog with cutaneous
area, proximal duodenum, and ileocolic junction. Mesen- pythiosis on a distal extremity.
teric lymphadenopathy is common but is usually caused by In cats, lesions associated with cutaneous pythiosis include
reactive hyperplasia rather than infection. Extension of dis- cervical, inguinal, or truncal subcutaneous masses; draining
ease into the mesenteric root often causes severe mesenteric nodular lesions or ulcerated plaque-like lesions on the tail-
lymphadenopathy, with the lymph nodes embedded in a head or extremities12; and periorbital and nasopharyngeal
single large, firm mass that is palpable in the cranial to mid lesions.13
670 SECTION 3  Fungal and Algal Diseases

Diagnosis
Laboratory Abnormalities
The most common laboratory abnormalities associated with
pythiosis are eosinophilia, anemia, hyperglobulinemia, and
hypoalbuminemia. Hypercalcemia has been described once.14

Imaging Findings
In dogs with gastrointestinal pythiosis, common radiographic
findings include poor abdominal detail and the presence of an
abdominal mass. Evidence of small bowel obstruction may also
be observed. In rare cases of esophageal pythiosis, thoracic
radiographs reveals increased soft tissue opacity in the region
of the esophagus and deviation of the trachea. Abdominal ultra-
sonography is the tool of choice for imaging animals suspected
to have gastrointestinal pythiosis and usually reveals severe seg-
mental thickening of the gastrointestinal tract and mesenteric
lymphadenopathy. In addition, Doppler ultrasonography may
identify the presence of vascular pathology (such as an aneu-
rysm) that can result from invasion of mesenteric vessels. In
the author’s practice, abdominal ultrasonography is a critical
tool for determining the location and extent of disease in dogs
with gastrointestinal pythiosis to determine the likelihood that
surgical resection could be performed successfully and to assess
prognosis. In addition, ultrasonography facilitates the acquisi-
tion of fine-needle aspirate specimens from affected segments of
the gastrointestinal tract and enlarged lymph nodes. In animals
with cutaneous pythiosis, computed tomography may provide
information about the extent of disease and may assist with sur-
gical planning when lesions are not located on an extremity.15

Microbiologic Tests
FIGURE 69-3  Cutaneous pythiosis in a 6-month-old female Labrador retriever.
(Courtesy Amy Grooters, Louisiana State University, Baton Rouge, LA.) Diagnostic assays currently available for pythiosis, lagenidiosis,
and zygomycosis in dogs and cats are described in Table 69-1.

TABLE 69-1
Diagnostic Assays Currently Available for Pythiosis, Lagenidiosis, and Zygomycosis in Dogs and Cats
Assay Specimen Type Target Performance
Cytologic exami- Aspirates of Broad, rarely septate Does not reliably differentiate among pythiosis, lagenidiosis,
nation ­affected tissues fungal hyphae with and zygomycosis.
tapered, rounded ends
Culture Biopsies of affected Pythium insidiosum, Special tissue handling and expertise required for P. insid-
tissues Lagenidium spp., or iosum culture (see text). The use of swab specimens or
zygomycetes aspirates may result in false negatives. Species identifica-
tion requires the use of molecular techniques.
Antibody serol- Serum Antibodies to P. insid- Assays that detect anti-Pythium antibodies allow early, non-
ogy iosum or Lagenidium invasive diagnosis of pythiosis in dogs. Positive test results
spp. correlate with active disease, and titers fall with successful
treatment. Assays that detect Lagenidium antibodies in
dogs are less specific and may be positive in animals with
other diseases.
Histopathology Biopsies of affected Broad, rarely septate Does not reliably differentiate among pythiosis, lagenidi-
tissues fungal hyphae with osis, and zygomycosis. Use of special stains may facilitate
tapered, rounded ends detection.
PCR assays Biopsies of affected P. insidiosum DNA Not widely available.
tissues
CHAPTER 69  Pythiosis, Lagenidiosis, and Zygomycosis 671

Cytologic Examination composed of epithelioid macrophages, plasma cells, multi-


Cytologically, pythiosis is characterized by pyogranulomatous nucleate giant cells, and fewer neutrophils and eosinophils are
and eosinophilic inflammation that can be visualized on impres- often observed. Organisms are typically found within areas
sion smears made from draining skin lesions or fine-needle of necrosis or at the center of granulomas. Vasculitis is occa-
aspirates of a thickened gastrointestinal wall or enlarged lymph sionally present. In gastrointestinal pythiosis, inflammation
nodes. Hyphae are observed occasionally in cytologic samples, centers on the submucosal and muscular layers rather than
and their morphologic appearance (broad, rarely septate with the mucosa and lamina propria. Therefore, the diagnosis of
tapered, rounded ends) in conjunction with a typical inflam- pythiosis may be missed if endoscopic biopsies that fail to
matory response can provide a tentative diagnosis of pythiosis, reach deeper tissues are submitted. Similarly, disease in ani-
lagenidiosis, or zygomycosis.14 mals with cutaneous pythiosis is typically found in the deep
dermis and subcutis, necessitating deep wedge biopsies rather
Culture than punch biopsies for optimal evaluation. Pythium insid-
Isolation of P. insidiosum from infected tissues is not dif- iosum hyphae are not routinely visualized on H&E-stained
ficult but does require specific specimen handling and culture sections but may be identified as clear spaces surrounded by
techniques. For best results, unrefrigerated tissue specimens a narrow band of eosinophilic material. Hyphae are readily
should be wrapped in a saline-moistened gauze sponge and visualized in sections stained with Gomori’s methenamine sil-
shipped at ambient temperature to arrive within 24 hours ver (GMS) but usually do not stain well with periodic acid–
at a laboratory that has experience with culture of patho- Schiff (PAS). They are wide (mean, 4 µm; range, 2 to 7 µm),
genic oomycetes. Small pieces of fresh, nonmacerated tissue have nonparallel walls, are infrequently septate, and occasion-
are placed directly on the surface of vegetable extract agar ally branch at right angles.7,8
supplemented with streptomycin and ampicillin (or an alter-
native selective medium) and incubated at 37°C.16 Mycelial Treatment and Prognosis
growth is typically observed within 12 to 24 hours. Isola-
tion of P. insidiosum from swabs of exudate collected from Aggressive surgical resection of all infected tissues with wide
draining skin lesions is generally unsuccessful. Because gen- margins is the treatment of choice for pythiosis. In animals
eration of the sexual reproductive structures that are neces- with cutaneous lesions that are confined to a single distal
sary for definitive morphologic classification of pathogenic extremity, amputation should be recommended unless there
oomycetes rarely occurs in the laboratory, identification of is evidence of regional lymph node infection. In patients
P. insidiosum isolates is based on species-­specific PCR ampli- with gastrointestinal pythiosis, segmental lesions should be
fication17 or rRNA gene sequencing. Although production resected with 5-cm margins whenever possible. Because mes-
of zoospores is an important supporting feature for the enteric lymph nodes are usually reactive rather than infected,
identification of pathogenic oomycetes, it is not specific for the presence of nonresectable mesenteric lymphadenopathy
P. insidiosum. should not dissuade the surgeon from pursuing resection of
a segmental bowel lesion. Enlarged regional lymph nodes
Molecular Diagnosis using the Polymerase Chain Reaction should, however, always be biopsied for prognostic informa-
A species-specific PCR assay has been used to identify P. insid- tion. In animals with cutaneous pythiosis, the surgeon should
iosum DNA in fresh, frozen, or paraffin-embedded tissues, as attempt to obtain skin margins of 5 cm and deep margins of
well as DNA extracted directly from cultured isolates.18 How- two fascial planes.15 Enlarged regional lymph nodes in dogs
ever, such assays are not widely available on a commercial with cutaneous pythiosis are often infected rather than reac-
basis. tive; therefore, they should be evaluated cytologically or his-
tologically before amputation or other aggressive resection is
Serologic Testing attempted.
A highly sensitive and specific ELISA for the detection of anti– Unfortunately, many dogs with pythiosis are not brought
P. insidiosum antibodies in dogs is currently available through to a veterinarian until late in the course of disease, when
the author’s laboratory at Louisiana State University.19 This complete excision is not possible. In addition, the anatomic
assay provides a means for early, noninvasive diagnosis and location of the lesion may prevent complete surgical excision
also allows response to therapy to be monitored. Follow- when gastrointestinal pythiosis involves the esophagus, gastric
ing complete surgical resection of infected tissues, a dramatic outflow tract, rectum, or mesenteric root, or when cutaneous
decrease in antibody levels is typically detected within 2 to 3 pythiosis involves the ventral thorax, base of the tail, or trunk.
months. In contrast, antibody levels remain high in animals Still, very aggressive surgery such as a partial gastrectomy14 or
that go on to develop clinical recurrence following surgical massive resection of cutaneous lesions combined with recon-
treatment. This same ELISA has been adapted for detection of structive techniques15 can sometimes result in a successful
anti–P. insidiosum antibodies in domestic and exotic cats, and outcome.
although case numbers are too small to generate strong esti- Local postoperative recurrence of pythiosis is common
mates of sensitivity and specificity, in the author’s experience (especially when wide surgical margins cannot be achieved)
they appear to be high. and can occur either at the site of resection or in regional
lymph nodes. For this reason, postoperative medical therapy
Pathologic Findings is often recommended when the surgeon is not confident that
Histologically, pythiosis is characterized by eosinophilic pyo- margins of at least 5 cm were achieved. In the author’s prac-
granulomatous inflammation, with multiple foci of necrosis tice, dogs with focal midjejunal lesions that are resected with
that are surrounded and infiltrated by neutrophils, eosino- wide margins and dogs without regional lymphadenopathy
phils, and macrophages. In addition, discrete granulomas that undergo amputation for distal extremity lesions are not
672 SECTION 3  Fungal and Algal Diseases

routinely treated with postoperative antifungal medication.


All other patients with pythiosis typically are treated with itra- Overview of Lagenidiosis
conazole (10 mg/kg PO q24h) and terbinafine (5 to 10 mg/kg First Described: 2003, Louisiana, USA, in dogs (Grooters and
PO q24h) for at least 2 to 3 months after surgical resection. others) 23
Unfortunately, if resection is incomplete, lesions often progress
Cause: Lagenidium spp.
despite medical therapy, and clinical signs recur within weeks
to months. Affected Hosts: Dogs
To monitor for recurrence, ELISA serology should be Geographic Distribution: Southeastern United States
performed at the time of surgery and 2 to 3 months later. In
Mode of Transmission: Likely penetration of damaged skin or
animals that have had a complete surgical resection and subse-
mucosa by motile zoospores
quently show no recurrence of disease, serum antibody levels
usually drop 50% or more within 3 months. If this occurs, Major Clinical Signs: Ulcerative, nodular, and mass-like cuta-
medical therapy can be discontinued. In the author’s experi- neous lesions with draining tracts (cutaneous form);
ence, surgery is curative in a majority of animals with a dis- pelvic limb edema; local, thoracic, or abdominal lymph-
tal limb lesion treated with amputation, or with a midjejunal adenopathy; rupture of infected great vessels in the abdo-
lesion that the surgeon believes was completely resected with men may result in hemoabdomen
good margins. Differential Diagnosis: Neoplasia, pythiosis, zygomycosis,
Medical therapy for nonresectable pythiosis is typically mycobacterial infections, actinomycosis, snake bite
unrewarding, likely because ergosterol (the target for most
currently available antifungal drugs) is generally lacking in the Human Health Significance: Lagenidium spp. infection is
oomycete cell membrane. Despite this fact, clinical and sero- acquired from the environment; infection is not transmit-
logic cures occur in some patients treated with a combination ted from affected animals to people.
of itraconazole (10 mg/kg PO q24h) and terbinafine (5 to 10
mg/kg PO q24h).20 Although the percentage of animals that
respond to this treatment is still low, the combination protocol LAGENIDIOSIS
anecdotally appears to be superior to itraconazole or ampho-
tericin B alone. Etiology and Epidemiology
Dogs with nonresectable gastrointestinal pythiosis are often
treated with anti-inflammatory doses of glucocorticoids in an Most species in the genus Lagenidium are pathogens of insects,
effort to palliate clinical signs and to decrease vomiting so that crustaceans, algae, and nematodes. The most well studied species,
oral antifungal drugs can be administered. Prednisone (1 mg/ Lagenidium giganteum, is a mosquito larval pathogen that was
kg/day) routinely causes improvement in clinical signs in the previously used as a biologic pesticide for control of mosquito
short term. Surprisingly, the author has observed complete and populations. Within the past 15 years, two novel oomycetes that
long-term resolution of gastrointestinal lesions (based on serial appear to belong to the genus Lagenidium have been isolated
ultrasonographic examinations and serologic testing) in a small from dogs with cutaneous lesions. The first of these species causes
number of dogs treated with prednisone alone. Although this is uniformly fatal dermatologic and disseminated disease in dogs in
certainly not recommended as a primary treatment for animals the southeastern United States.23 The second pathogenic Lagen-
with resectable lesions, it is a reasonable option in animals with idium species is less common than the first and causes a chronic
nonresectable gastrointestinal lesions, especially when financial ulcerative nodular dermatopathy that has a prolonged course and
concerns preclude the use of antifungal medication. The author does not appear to extend beyond local tissues. Although strong
has not observed this effect of prednisone in dogs with cutaneous antigenic and molecular similarities suggest that the first canine
pythiosis. pathogen is closely related to L. giganteum, differences in their
An immunotherapy product derived from antigens of molecular and in vitro growth characteristics suggest that they are
P. insidiosum has been used successfully to treat pythiosis in likely distinct species. Sporulation and infectivity for these patho-
horses and people.21,22 Unfortunately, although controlled tri- gens is thought to be similar to that associated with P. insidiosum
als have not been completed, the efficacy of this product in dogs and L. giganteum, causing infection through production of motile
appears to be poor, and clinical improvement has not been aquatic zoospores that adhere to and encyst in damaged tissues.
observed in any of the author’s patients. The epidemiologic features of lagenidiosis are similar in many
respects to those associated with cutaneous pythiosis. Affected
Public Health Aspects animals are typically young to middle-aged dogs that live in the
southeastern United States. Although most affected dogs have
Human infections caused by P. insidiosum are rare. Infections lived in Florida or Louisiana, cases in Texas, Tennessee, Ala-
are acquired from the environment and have resulted in kerati- bama, Georgia, South Carolina, Maryland, Virginia, Indiana,
tis, periorbital cellulitis, and arteritis of the lower extremities. and Illinois have also been identified. A number of infected dogs
Most cases have been reported from Thailand, and almost all have had frequent exposure to lakes or ponds. None have had
patients have thalassemia. Extremely rare reports of the disease historical evidence of immunocompromise or were treated with
exist from humans in North, South, and Central America, Aus- immunosuppressive therapy before development of the infection.
tralia, and other parts of Asia, many of whom are apparently
healthy. Although there is no evidence to suggest that transmis- Clinical Features
sion between mammalian hosts is possible, routine precautions
(such as wearing gloves when handling infected tissues or exu- Dogs with Lagenidium infection are typically evaluated for pro-
date) should be followed. gressive, multifocal or focal, cutaneous or subcutaneous lesions
CHAPTER 69  Pythiosis, Lagenidiosis, and Zygomycosis 673

Laboratory Abnormalities
Laboratory abnormalities are often absent in dogs with lagen-
idiosis; when present, they most often include hyperglobulin-
emia and eosinophilia. Hypercalcemia has been associated with
Lagenidium infection in one dog with cutaneous lesions on all
four limbs and severe infection and enlargement of popliteal,
inguinal, and superficial cervical lymph nodes.

Imaging Findings
Radiographic imaging of the thorax and sonographic imaging
of the abdomen are essential parts of the diagnostic evaluation
in dogs suspected to have lagenidiosis because of the potential
for occult lesions in the thorax or abdomen. These may include
solitary pulmonary nodules; sublumbar, inguinal, and medial
iliac lymphadenopathy; thickening and invasion of the wall
of the aorta or caudal vena cava (sometimes with associated
aneurysm); and retroperitoneal or epaxial masses. Thoracic and
abdominal lesions have not been observed in dogs infected with
FIGURE 69-4  Tailhead mass caused by lagenidiosis in a 9-year-old female spayed
the less aggressive Lagenidium species.
mixed breed dog. Note the extensive tissue necrosis. (Courtesy Amy Grooters, Louisiana
State University, Baton Rouge, LA.) Microbiologic Tests
Cytologic Examination
that involve the extremities, mammary region, perineum, or Cytologic examination of lymph node aspirates or impression
trunk.23 These lesions may appear as firm dermal or subcuta- smears made from draining lesions in dogs with lagenidiosis
neous nodules, or as ulcerated, thickened, edematous areas of reveal pyogranulomatous to eosinophilic inflammation with or
deep cellulitis with regions of necrosis and numerous draining without broad, poorly septate hyphae.
tracts (Figure 69-4). As with cutaneous pythiosis, skin lesions in
dogs with lagenidiosis tend to be progressive, locally invasive, Culture
and poorly responsive to medical therapy. The diagnosis of lagenidiosis is best made by culture followed by
In dogs infected with the more aggressive species of Lagen- PCR and rRNA gene sequencing, which not only provides a defini-
idium, regional lymphadenopathy is often noted and may tive diagnosis but is also the only tool currently available that differ-
occur in the absence of obvious cutaneous lesions. Unlike dogs entiates between the two pathogenic species. Isolation techniques
with cutaneous pythiosis, these dogs typically have occult for Lagenidium spp. are similar to those described for P. insidio-
lesions in the thorax or abdomen, which may involve the sum, but with peptone-yeast-glucose (PYG) agar. For best results,
great vessels, sublumbar and/or inguinal lymph nodes, lung, small pieces of fresh, nonmacerated tissue are placed directly on
pulmonary hilus, and cranial mediastinum. Animals with the surface of the agar and incubated at 37°C. Growth is typically
great vessel or sublumbar lymph node involvement usually observed within 24 to 48 hours. Because production of the sexual
have cutaneous or subcutaneous lesions on the pelvic limbs reproductive structures necessary for morphologic classification
and often develop pelvic limb edema. Sudden death caused by has not yet been possible, definitive identification of Lagenidium
great vessel rupture and associated hemoabdomen may occur species is currently based on rRNA gene sequencing. Although
in these animals. genus-specific PCR assays have been used to identify Lagenidium
In dogs infected with the less aggressive species of Lagen- DNA extracted from cultured isolates or from infected tissue speci-
idium, lesions tend to be locally invasive but rarely extend mens,18 the clinical importance of differentiating between the two
beyond cutaneous and subcutaneous tissues. Distant lesions in pathogenic species in regard to prognosis and treatment makes cul-
the thorax and abdomen have not been identified, and the clini- ture and rRNA gene sequencing the preferred technique.
cal course appears to be chronic and slowly progressive; some
dogs have lesions that are somewhat stable for several years. Serologic Testing
In dogs with supportive clinical signs and histologic findings, immu-
Diagnosis noblot serology for the detection of anti-Lagenidium antibodies
in canine serum can suggest lagenidiosis but must be interpreted
Because of its clinical, epidemiologic, and histologic similari- in conjunction with results of serologic testing for P. insidiosum
ties to pythiosis, lagenidiosis is often misdiagnosed as pythio- infection because of the potential for cross-reactivity in serum from
sis during routine histologic evaluation. In addition, because dogs with pythiosis. In addition, the author has observed nonspe-
many pathologists are more familiar with the zygomycetes than cific anti-Lagenidium seroreactivity in dogs with other fungal or
the oomycetes, lagenidiosis may also be misdiagnosed as zygo- nonfungal infections. Therefore, based on currently available data,
mycosis. Although there are subtle differences in hyphal size, serology alone should not be used as a basis for the diagnosis of
morphology, and distribution among these three infections, his- lagenidiosis. Serology is not helpful for differentiating between
tologic lesions associated with lagenidiosis are still often labeled infection by the two canine pathogenic species of Lagenidium.
as “suspected pythiosis” or “suspected zygomycosis” during
routine evaluation. For this reason, clinicians should always Pathologic Findings
attempt to confirm a suspected histologic diagnosis with serol- Histologically, lagenidiosis shares many characteristics with
ogy, culture, or molecular diagnostic assays. pythiosis and zygomycosis, including pyogranulomatous and
674 SECTION 3  Fungal and Algal Diseases

Public Health Aspects


Infections caused by Lagenidium species are acquired from
the environment. Although there is no evidence to suggest that
transmission between mammalian hosts is possible, routine pre-
cautions (such as wearing gloves when handling infected tissues
or exudate) should be followed.

Overview of Zygomycosis
(Entomophthoramycosis and Mucormycosis)
First Described: Reports of zygomycosis in humans date back
to the 1800s.
Causes: Basidiobolus and Conidiobolus spp. (kingdom Fungi,
order Entomophthorales); Rhizopus, Absidia, Mucor spp.,
and others (kingdom Fungi, order Mucorales)
Affected Hosts: Dogs, cats, humans, horses, sheep, other
FIGURE 69-5  Broad, thick-walled infrequently septate hyphae associated with gran- mammalian species
ulomatous vasculitis in a lymph node from a dog infected with the more aggressive species
of Lagenidium. (Courtesy Amy Grooters, Louisiana State University, Baton Rouge, LA.) Geographic Distribution: Worldwide
Mode of Transmission: Inhalation, ingestion, or cutaneous
exposure to organisms in soil and decaying organic matter
Major Clinical Signs: Entomophthoramycoses are associated
eosinophilic inflammation associated with broad, irregularly with chronic, slowly progressive subcutaneous, retrobul-
branching, infrequently septate hyphae with nonparallel walls. bar, or nasal infections in immunocompetent animals;
Multinucleated giant cells and plasma cells are commonly mucormycoses are associated with systemic (respiratory
present. In contrast to P. insidiosum, Lagenidium spp. hyphae or gastrointestinal) or cutaneous-subcutaneous infec-
are often visible on H&E-stained sections. On GMS-stained tions in immunocompromised animals.
sections, numerous broad, thick-walled, irregularly sep-
tate hyphae are easily recognized (Figure 69-5). Lagenidium Differential Diagnosis: Neoplasia, pythiosis, lagenidiosis,
hyphae typically demonstrate a great deal of variability in size, other deep mycoses (especially blastomycosis), mycobac-
but in general are larger than P. insidiosum hyphae, ranging terial infections, actinomycosis
from 7 to 25 µm in diameter, with an average of 12 µm for Human Health Significance: Disease in humans results from
the more aggressive pathogen and 7.5 µm for the less aggres- exposure to organisms in the environment, and direct
sive pathogen. In some sections, hyphae appear as round or transmission between animals has not been reported.
bulbous structures, and right-angle branching is occasionally
observed. A scant to thin eosinophilic sleeve may be noted
around the hyphae.

Treatment and Prognosis ZYGOMYCOSIS


As with pythiosis, aggressive surgical resection of infected tis- Etiology and Epidemiology
sues is the treatment of choice for lagenidiosis when disease is
confined to a resectable cutaneous lesion. Unfortunately, the The term “zygomycosis” refers to infections caused by fungi
vast majority of dogs infected with the more common (and more in the class Zygomycetes, which includes pathogenic fungi of
aggressive) Lagenidium pathogen have nonresectable disease the genera Basidiobolus and Conidiobolus (order Entomoph-
in the thorax, abdomen, or regional lymph nodes by the time thorales) and the genera Rhizopus, Absidia, Mucor, and oth-
the initial diagnosis is made, and in the author’s experience, the ers (order Mucorales). In human and veterinary patients, the
disease is routinely fatal. In dogs infected with the less aggres- Entomophthorales typically cause chronic localized infections
sive species, surgery that achieves 3- to 5-cm margins is often in subcutaneous tissue or nasal submucosa of immunocom-
curative. Because the species of Lagenidium present is often not petent patients that usually reside in tropical or subtropical
known early in the course of diagnostic evaluation, any dog sus- locations (entomophthoramycoses), whereas the Mucorales
pected of having lagenidiosis should be evaluated with thoracic tend to cause acute, rapidly progressive disease in debilitated
radiography and abdominal ultrasonography before surgical or immunocompromised individuals worldwide (mucormyco-
resection of cutaneous lesions is attempted. As with pythiosis, ses). Basidiobolus spp. and Conidiobolus spp. cause cutane-
medical therapy for lagenidiosis is usually ineffective. How- ous lesions in dogs that are grossly and histologically similar
ever, treatment with a combination of itraconazole (10 mg/kg to those caused by P. insidiosum and Lagenidium spp. Cul-
PO q24h) and terbinafine (5 to 10 mg/kg PO q24h) along with ture-confirmed infections caused by pathogens in the order
repeated aggressive surgical resection was curative in one dog Mucorales have not been well documented in dogs but have
with recurrent multifocal cutaneous lesions caused by the less been described in a small number of cats. Unfortunately, clini-
aggressive pathogen. cal and pathologic information about zygomycosis in small
CHAPTER 69  Pythiosis, Lagenidiosis, and Zygomycosis 675

A B
FIGURE 69-6  A, Nasopharyngeal Conidiobolus spp. infection in a 4-year-old male neutered German shepherd dog. B, Note the severe swelling of the nose and muzzle and ulceration
of the nasal planum. (Courtesy Amy Grooters, Louisiana State University, Baton Rouge, LA.)

animal patients is lacking because disease is uncommon to In cats, culture-confirmed cases of zygomycosis are sparse
rare, and establishment of a definitive diagnosis has histori- and have been limited to single reports of cats infected with
cally required tissue biopsy and culture. As a result, small ani- fungi in the order Mucorales. Rhizomucor was identified as
mal patients with zygomycosis often go undifferentiated from the cause of duodenal perforation in a 7-month-old cat,28 and
those with pythiosis. Cokeromyces recurvatus was isolated from abdominal fluid in
Basidiobolus and Conidiobolus species are saprophytes a 16-year-old cat following jejunal perforation caused by lym-
commonly found in soil and decaying plant matter. Cutaneous phosarcoma.29 In addition, a subcutaneous mass caused by a
infection with Basidiobolus or Conidiobolus likely occurs by Mucor species on the dorsum of the nose of a 14-year-old cat
percutaneous inoculation of spores via minor trauma or insect was treated successfully with posaconazole.30
bites. Infection may also result from inhalation or ingestion of
spores. Diagnosis
Clinical Features Because of their histologic similarities, zygomycosis is often
confused with pythiosis (which is much more common) when
In dogs, humans, horses, sheep, and other mammalian spe- tissue biopsies are evaluated. Unfortunately, there are no sero-
cies, conidiobolomycosis occurs most often as a nasopha- logic, immunohistochemical, or molecular techniques that are
ryngeal infection with or without local dissemination into routinely available for the diagnosis of zygomycosis, making
tissues of the face, retropharyngeal region, and retrobulbar identification of infected animals reliant on fungal culture of
space. Manifestations of infection in dogs may include nasal fresh tissues. As a result, a definitive diagnosis of zygomycosis
or facial swelling or deformity, nasal discharge, ulceration is often elusive.
of the nasal planum or hard palate, exophthalmos, chemo-
sis, ocular discharge, and sometimes skin lesions near the Laboratory and Imaging Abnormalities
eye (Figure 69-6). In animals with retrobulbar disease that Laboratory abnormalities are not well described in animals
extends into the brain, neurologic signs may occur. Conid- with zygomycosis. Diagnostic imaging in the form of computed
iobolus infection has been described in a single dog as a cause tomography often provides important information about the
of multifocal nodular draining subcutaneous lesions and location and extent of disease in animals with nasopharyngeal
regional lymphadenopathy24 and as a cause of pneumonia in and retrobulbar lesions caused by Conidiobolus spp. In addi-
a dog that was receiving chemotherapy.25 Basidiobolomyco- tion, ocular ultrasonography may help to characterize the extent
sis is a rare cause of ulcerative skin lesions in dogs and has of disease in dogs with retrobulbar lesions.
also been reported as a cause of respiratory disease in a dog.26
Disseminated Basidiobolus infection involving the gastroin- Cytologic and Histologic Findings
testinal tract and other abdominal organs has been described The cytologic and histologic features of zygomycosis are simi-
in two dogs.7,27 lar to those associated with pythiosis and lagenidiosis. On
676 SECTION 3  Fungal and Algal Diseases

Treatment and Prognosis


Recommendations for the treatment of zygomycosis are not
straightforward because attempted therapy has only been
described in a few patients with culture-confirmed diagnoses.
Although anecdotal information as well as a small number of
cases in the literature suggest that cutaneous zygomycosis may
be less aggressive than cutaneous pythiosis or lagenidiosis, pro-
gression of lesions and sometimes even dissemination despite
treatment have also been observed in zygomycete-infected dogs.
The author’s current recommendation for dogs with nasopha-
ryngeal conidiobolomycosis is treatment with itraconazole
(10 mg/kg PO q24h) for at least 6 months. Recurrence is com-
mon after medication is discontinued, so a prolonged course
is usually prescribed. Newer oral azoles such as posaconazole
FIGURE 69-7  Conidiobolus spp. hyphae in a tissue biopsy from an ulcerated palate would also be reasonable choices but are significantly more
lesion in a 3-year-old female boxer. Note the large amount of amorphous eosinophilic expensive. Cutaneous zygomycosis in small animal patients
material (sleeve) surrounding the hyphal segment; H&E stain. (Courtesy Amy Grooters, should be treated with aggressive surgical resection of infected
Louisiana State University, Baton Rouge, LA.) tissues whenever possible, followed by itraconazole therapy
for 2 to 3 months. If resection is not possible, treatment with
GMS-stained sections, hyphae are broad, thin-walled, and itraconazole, posaconazole, or lipid-complexed amphotericin B
occasionally septate. The histologic hallmark of zygomycosis should be recommended.
is the presence of a wide (2.5 to 25 µm) eosinophilic sleeve that
surrounds the hyphae and makes them easily located on H&E- Public Health Aspects
stained sections (Figure 69-7). This finding helps to differen-
tiate zygomycosis from pythiosis and lagenidiosis, in which Zygomycosis in human patients is rare. Infections are acquired
eosinophilic sleeves tend to be thin or absent. The hyphal diam- from the environment. Although there is no evidence to suggest
eter is also significantly larger for Basidiobolus spp. (mean 9 that transmission between mammalian hosts occurs, routine
µm; range, 5-20 µm) and Conidiobolus spp. (mean 8 µm; range, precautions (such as wearing gloves when handling infected tis-
5-13 µm) than for P. insidiosum (mean, 4 µm; range, 2-7 µm).31 sues or exudate) should be followed.

CASE EXAMPLE Musculoskeletal: The dog was underweight with a body


condition score of 3/9.
Cardiovascular and Respiratory: Strong femoral pulses. No
Signalment: “Jasper,” an 18-month-old intact male Labrador murmurs or arrhythmias were auscultated. Respiratory rate
retriever from central Louisiana and effort were normal.
History: Jasper was evaluated for a 1-month history of Gastrointestinal and Genitourinary: The patient showed
frequent vomiting and persistent diarrhea and a 2-week signs of discomfort during abdominal palpation; a large,
history of intermittent anorexia. In addition, the owner firm, tubular mass was palpated in the midabdomen. Mild
reported that the dog had lost approximately 6 kg. The prostatomegaly was detected on rectal examination.
diarrhea was watery and yellowish-brown and was produced Lymph nodes: All palpable peripheral lymph nodes were
in large quantity; hematochezia had not been noted. A fecal normal in size and shape.
flotation and heartworm antigen test performed by the Laboratory Findings:
referring veterinarian 1 week before the dog was evaluated CBC:
at the author’s institution had been negative. Treatment HCT 33.7% (37%-55%)
with metronidazole (17 mg/kg PO q12h) and a change to MCV 61.7 fL (62-77 fL)
an easily digestible prescription diet had failed to improve MCHC 20.8 g/dL (32-37 g/dL)
the vomiting and diarrhea. The patient was a field trial dog WBC 10,600 cells/µL (8000-14,500 cells/µL)
used for duck and dove hunting in central Louisiana. He Neutrophils 6200 cells/µL (3000-11,500 cells/µL)
frequently worked in rice fields and occasionally in bodies Lymphocytes 1700 cells/µL (1000-4800 cells/µL)
of fresh water and was generally housed in an outdoor Monocytes 1100 cells/µL (100-1400 cells/µL)
kennel. Platelets 254,000/µL (220,000-600,000 platelets/µL).
Physical Examination: Serum Chemistry Profile:
Body Weight: 30 kg. Sodium 142 mmol/L (140-153 mmol/L)
General: Bright, alert, and responsive. Body temperature = Potassium 4.4 mmol/L (3.8-5.5 mmol/L)
102.7°F (39.2°C), HR = 110 beats/min, RR = 20 breaths/min, Chloride 110 mmol/L (107-115 mmol/L)
mucous membranes pink, CRT ~ 2 s. Bicarbonate 20 mmol/L (17-27 mmol/L)
Eyes, ears, nose, and throat: No significant abnormalities Phosphorus 4.6 mg/dL (3.4-6.3 mg/dL)
were noted. Calcium 9.1 mg/dL (9.4-11.4 mg/dL)
CHAPTER 69  Pythiosis, Lagenidiosis, and Zygomycosis 677

BUN 11 mg/dL ( 8-22 mg/dL)


Creatinine 0.7 mg/dL (0.5-1.7 mg/dL)
Glucose 96 mg/dL (80-115 mg/dL) Jejunum
Total protein 5.7 g/dL (5.8-7.5 g/dL)
Albumin 2.0 g/dL (2.6-4.2 g/dL)
Globulin 3.7 g/dL (2.5-4.0 g/dL)
ALT 34 U/L (0-60 U/L)
AST 22 U/L (0-50 U/L)
ALP 13 U/L (0-100 U/L)
Creatine kinase 134 U/L (0-200 U/L)
Cholesterol 158 mg/dL (150-240 mg/dL)
Total bilirubin 0.2 mg/dL (0-0.4 mg/dL).
Urinalysis: SGr 1.041, pH 7.5, 3+ protein, 3+ bilirubin, negative
hemoglobin, negative glucose, negative ketones, 0-5 WBC/
HPF, 0 RBC/HPF, many lipid droplets.
Imaging Findings:
Plain Abdominal Radiographs: There was a generalized
A Dist 1.16 cm
loss of serosal detail in the abdomen. Gas was present in
the gastrointestinal tract, but there was no evidence of
abnormal intestinal tract dilation.
Abdominal Ultrasound: A severely thickened segment of Mes L Node
small bowel could be visualized extending from the cranial
to the caudal abdomen. The wall of this bowel segment
measured 1.1 cm in thickness and lacked normal layering
(Figure 69-8, A). Mesenteric lymph nodes were enlarged and
heterogenous (Figure 69-8, B). There was a small amount of
free abdominal fluid.
Cytology Findings: Cytologic evaluation of ultrasound-
guided mesenteric lymph node aspirates revealed reactive
lymphoid hyperplasia. Examination of ultrasound-guided
aspirates of the thickened segment of bowel showed that
the cellularity was too low for cytologic evaluation.
Serology Findings: ELISA serology for anti–Pythium Dist 4.11 cm
Dist 1.79 cm
insidiosum antibodies: Positive at 69%. Results of this assay are
expressed as percent positivity in comparison with a strong
B
positive control sample, with values > 40% positivity having
been shown to be 100% specific for pythiosis in dogs. Values FIGURE 69-8  Sonographic images of the abdomen of the dog described in the
in healthy dogs range from 3% to 15%.19 case example. Note the severe thickening and loss of layering in the wall of the jejunum
Diagnosis: Jejunal pythiosis. (A) and the enlarged, heterogenous appearance of the mesenteric lymph node (B).
Treatment: Abdominal exploratory revealed a 50-cm segment
of the mid-jejunum that was severely thickened and firm.
This segment was resected with 5-cm margins. After an and had gained 3 kg body weight. Reevaluation of anti–
uneventful postoperative recovery, the dog’s vomiting, Pythium insidiosum antibody serology at that time yielded
diarrhea, and anorexia resolved quickly. He was treated a percent positivity of 20%. Because the rapid decrease in
with itraconazole (10 mg/kg PO q24h) and terbinafine (8.3 antibody levels from 69% to 20% was strongly suggestive
mg/kg PO q24h) for 2 months in an effort to decrease the of a surgical cure of gastrointestinal pythiosis, oral
chance of recurrence of disease at the site of resection. Liver antifungal medications were discontinued. Reevaluation
enzyme activities and serum bilirubin concentration were of anti–Pythium insidiosum antibody serology 2 years
monitored during this time and remained within reference later yielded results within the range observed in healthy
intervals. dogs (6%).
Histopathologic Findings: Both the resected segment of Comments: Although most dogs with gastrointestinal
jejunum as well as jejunal and ileocolic lymph nodes were pythiosis have nonresectable lesions, those with mid-
submitted for histologic examination. Results revealed jejunal lesions can often be surgically cured. Preoperative
severe transmural eosinophilic pyogranulomatous enteritis abdominal ultrasound is essential since it helps to
and lymphoid hyperplasia. In the jejunal lesion, wide (3- to determine whether or not a gastrointestinal lesion might be
8-µm), poorly septate hyphae with nonparallel walls were surgically resectable. Because medical therapy alone is often
visualized on GMS-stained sections. No organisms were unsuccessful for the treatment of gastrointestinal pythiosis,
observed in the lymph node sections. surgery should be pursued unless there is clear evidence
Follow-up: At reevaluation 2 months after resection of the that lesions are nonresectable. Postoperative medical
jejunal lesion, the dog remained free of clinical signs therapy was probably not necessary in the case described

Continued
678 SECTION 3  Fungal and Algal Diseases

here, since the postoperative clinical course and follow-up the resection and lymph node histology shows hyperplasia
serology results strongly supported a surgical cure. In the rather than infection. In such cases, medical treatment with
author’s experience, most dogs with a midjejunal lesion are itraconazole and terbinafine can be offered as optional but
surgically cured when the surgeon is confident that greater likely unnecessary therapy for 2 months until follow-up
than 5-cm margins of healthy tissue were obtained with serology is performed.

SUGGESTED READINGS 17. Grooters AM, Gee MK. Development of a nested PCR assay for
the detection and identification of Pythium insidiosum. J Vet Intern
Chayakulkeeree M, Ghannoum MA, Perfect JR. Zygomycosis: the Med. 2002;16:147-152.
re-emerging fungal infection. Eur J Clin Microbiol Infect Dis. 18. Znajda NR, Grooters AM, Marsella R. PCR-based detection of
2006;25(4):215-229. Pythium and Lagenidium DNA in frozen and ethanol-fixed animal
Grooters AM. Pythiosis and lagenidiosis. In: Bonagura JD, ed. Kirk’s tissues. Vet Dermatol. 2002;13:187-194.
Current Veterinary Therapy XIV. St. Louis, MO: Saunders; 2008. 19. Grooters AM, Leise BS, Lopez MK, et al. Development and evalua-
Grooters AM. Pythiosis, lagenidiosis, and zygomycosis in small animals. tion of an enzyme-linked immunosorbent assay for the serodiagno-
Vet Clin North Am Small Anim Pract. 2003;33:695-720. sis of pythiosis in dogs. J Vet Intern Med. 2002;16:142-146.
20. Hummel J, Grooters A, Davidson G, et al. Successful management
REFERENCES of gastrointestinal pythiosis in a dog using itraconazole, terbin-
1. Kwon-Chung KJ. Phylogenetic spectrum of fungi that are patho- afine, and mefenoxam. Med Mycol. 2011;49:539-542.
genic to humans. Clin Infect Dis. 1994;19(suppl 1):S1-S7. 21. Hubert JD, Grooters AM. Treatment of equine pythiosis. Compend
2. Grooters AM. Pythiosis, lagenidiosis, and zygomycosis in small Contin Ed Pract Vet. 2002;13:187-194.
animals. Vet Clin North Am Small Anim Pract. 2003;33:695-720. 22. Wanachiwanawin W, Mendoza L, Visuthisakchai S, et  al. Effi-
3. Smith F. The pathology of bursattee. Vet J. 1884;19:16-17. cacy of immunotherapy using antigens of Pythium insidio-
4. Gaastra W, Lipman LJ, De Cock AW, et al. Pythium insidiosum: an sum in the treatment of vascular pythiosis in humans. Vaccine.
overview. Vet Microbiol. 2010;146:1-16. 2004;22:3613-3621.
5. Mendoza L, Hernandez F, Ajello L. Life cycle of the human and 23. Grooters AM, Hodgin EC, Bauer RW, et  al. Clinicopathologic
animal oomycete pathogen Pythium insidiosum. J Clin Microbiol. findings associated with Lagenidium sp. infection in six dogs:
1993;31:2967-2973. initial description of an emerging oomycosis. J Vet Intern Med.
6. Berryessa NA, Marks SL, Pesavento PA, et al. Gastrointestinal pythio- 2003;17:637-646.
sis in 10 dogs from California. J Vet Intern Med. 2008;22:1065-1069. 24. Hillier A, Kunkle GA, Ginn PE, et al. Canine subcutaneous zygo-
7. Miller RI. Gastrointestinal phycomycosis in 63 dogs. J Am Vet mycosis caused by Conidiobolus sp.: a case report and review
Med Assoc. 1985;186:473-478. of Conidiobolus infections in other species. Vet Dermatol.
8. Foil CSO, Short BG, Fadok VA, et  al. A report of subcutaneous 1994;5:205-213.
pythiosis in five dogs and a review of the etiologic agent Pythium 25. Hawkins EC, Grooters AM, Cowgill ES, et  al. Treatment of
spp. J Am Anim Hosp Assoc. 1984;20:959-966. Conidiobolus sp. pneumonia with itraconazole in a dog receiv-
9. Fischer JR, Pace LW, Turk JR, et al. Gastrointestinal pythiosis in ing immunosuppressive therapy. J Vet Intern Med. 2006;20:
Missouri dogs: eleven cases. J Vet Diagn Invest. 1994;6:380-382. 1479-1482.
10. Patton CS, Hake R, Newton J, et al. Esophagitis due to Pythium insid- 26. Greene CE, Brockus CW, Currin MP, et al. Infection with Basid-
iosum infection in two dogs. J Vet Intern Med. 1996;10:139-142. iobolus ranarum in two dogs. J Am Vet Med Assoc. 2002;221:
11. Rakich PM, Grooters AM, Tang KN. Gastrointestinal pythiosis in 528-532:500.
two cats. J Vet Diagn Invest. 2005;17:262-269. 27. Miller RI, Turnwald GH. Disseminated basidiobolomycosis in a
12. Thomas RC, Lewis DT. Pythiosis in dogs and cats. Compend Con- dog. Vet Pathol. 1984;21:117-119.
tin Ed Pract Vet. 1998;20:63-74. 28. Cunha SC, Aguero C, Damico CB, et  al. Duodenal perfora-
13. Bissonnette KW, Sharp NJ, Dykstra MH, et al. Nasal and retrobul- tion caused by Rhizomucor species in a cat. J Feline Med Surg.
bar mass in a cat caused by Pythium insidiosum. J Med Vet Mycol. 2011;13:205-207.
1991;29:39-44. 29. Nielsen C, Sutton DA, Matise I, et  al. Isolation of Cokeromyces
14. LeBlanc CJ, Echandi RL, Moore RR, et al. Hypercalcemia associated recurvatus, initially misidentified as Coccidioides immitis, from
with gastric pythiosis in a dog. Vet Clin Pathol. 2008;37:115-120. peritoneal fluid in a cat with jejunal perforation. J Vet Diagn Invest.
15. Thieman KM, Kirkby KA, Flynn-Lurie A, et  al. Diagnosis and 2005;17:372-378.
treatment of truncal cutaneous pythiosis in a dog. J Am Vet Med 30. Wray JD, Sparkes AH, Johnson EM. Infection of the subcutis of the
Assoc. 2011;239:1232-1235. nose in a cat caused by Mucor species: successful treatment using
16. Grooters AM, Whittington A, Lopez MK, et  al. Evaluation of posaconazole. J Feline Med Surg. 2008;10:523-527.
microbial culture techniques for the isolation of Pythium insidio- 31. Miller RI, Campbell RS. The comparative pathology of equine
sum from equine tissues. J Vet Diagn Invest. 2002;14:288-294. cutaneous phycomycosis. Vet Pathol. 1984;21:325-332.
CHAPTER 70

Protothecosis
Jane E. Sykes

may not inactivate Prototheca.2 Prototheca spp. also colonize


Overview of Protothecosis the skin and gastrointestinal and respiratory tracts of humans.3
First Described: Prototheca spp. was first isolated from tree The algae are round to oval, 5 to 30 µm in diameter, have
slime in the 1880s. Naturally occurring protothecosis a thick cell wall, and reproduce asexually by endosporulation
was not described until 1952, in cattle (mastitis). Canine (internal division), which results in a sporangium that con-
disease was reported in 1969, 5 years after the first tains up to multiple sporangiospores. Rupture of the sporan-
description of human protothecosis (Van Kruiningen and gium releases sporangia into tissues, and the cycle continues.
othes).1 Although the classification of Prototheca species is still in flux,
six species are currently recognized: Prototheca zopfii, Proto-
Cause: Prototheca spp. (lower algae, Chlorophyceae)
theca wickerhamii, Prototheca blaschkeae, Prototheca stag-
Affected Hosts: Dogs, cats, cattle, humans, other domestic nora, Prototheca ulmea, and Prototheca cutis (Table 70-1).4
and wild animal species Only P. zopfii and P. wickerhamii have been identified as
Geographic Distribution: Worldwide except Antarctica, but causes of disease in dogs and cats. Two genotypes of P. zopfii
especially in warm, humid climates and where there is are recognized. P. zopfii genotype 1 is a weakly pathogenic
organic matter with a high water content variant and has been isolated from a minority of protothecal
mastitis cases in dairy cows.4,5 In contrast, P. zopfii genotype 2
Mode of Transmission: Cutaneous inoculation of organisms may be the most virulent Prototheca variant. It causes the vast
into tissues, or possibly systemic invasion in the face majority of bovine Prototheca mastitis cases worldwide and
of immunosuppression by organisms that have been most protothecosis in dogs.4 P. wickerhamii is often associ-
ingested or colonize the intestinal tract ated with cutaneous disease and has to date been isolated from
Major Clinical Signs: Cutaneous nodules or masses or sys- all affected cats, some dogs, and almost all human patients
temic illness with weight loss, blindness due to ocular (see Table 70-1). In dogs, disease caused by P. wickerhamii
involvement, polyuria and polydipsia, hematochezia, may follow a less aggressive clinical course than that caused by
vomiting, lameness, neurologic signs P. zopfii genotype 2.6
Differential Diagnoses: Chlorellosis (a rare algal disease), a Protothecosis is an uncommon and sporadic disease of dogs
variety of fungal diseases (e.g., histoplasmosis, cryptococ- and is rare in cats. In dogs, protothecosis is usually a serious
cosis, sporotrichosis), intestinal pythiosis, neoplasia (espe-
cially round cell neoplasia), toxoplasmosis, neosporosis,
granulomatous colitis of boxer dogs, inflammatory bowel TABLE 70-1
disease
Human Health Significance: Human infections occur as
Major Species (and Genotypes) of Prototheca and
a result of exposure to organisms in the environment Their Clinical Relevance
and are often associated with underlying immune
compromise. Host Species
Species Infected Clinical Manifestations
P. zopfii Cattle Mastitis (low
­genotype 1 ­pathogenicity)
Etiology and Epidemiology P. zopfii Dogs, humans, Mastitis (cattle),
­genotype 2 cattle, other ­disseminated infections
Protothecosis is an uncommon cutaneous or systemic disease (dogs)
caused by Prototheca species, which are unicellular algae. Pro- P. wickerhamii Dogs, cats Cutaneous disease
totheca spp. lack chlorophyll, and as a result they are dependent
P. blaschkeae Cattle, humans Mastitis (cattle), ­cutaneous
on a saprophytic lifestyle. Although ubiquitous in nature, they
disease (humans)
are especially prevalent in warm, humid climates (e.g., southern
and southeastern United States, northeastern Australia, south- P. stagnora Nonpathogenic NA
ern continental Europe, Japan) in aqueous environments where P. ulmea Nonpathogenic NA
decaying organic matter is present. They have been isolated from P. cutis Humans Cutaneous disease
tree slime, sewage, fresh and saltwater sources, fish tanks, soil,
animal excrement, and foodstuffs. Routine water chlorination NA, Not applicable.

679
680 SECTION 3  Fungal and Algal Diseases

disseminated disease, but localized cutaneous disease occurs but rectal temperature may also be normal. Ocular involvement
occasionally. Most affected dogs do not have a history of may be manifested by blindness or evidence of conjunctivitis,
immunosuppressive drug therapy or illness. Boxer dogs and col- hyphema, uveitis, cataract formation, panophthalmitis, or granu-
lies may be predisposed,6 possibly secondary to an underlying lomatous chorioretinitis, often with retinal detachment. Arrhyth-
genetic immunodeficiency, although a variety of other small- mias with pulse deficits may be present in dogs with protothecal
and large-breed dogs also can be affected. Occasionally proto- myocarditis.7 Neurologic signs include obtundation, blindness,
thecosis occurs in dogs in conjunction with infection by other deafness, ataxia, abnormal placing reactions, circling, head tilt,
opportunistic pathogens such as Neospora,7 which supports the nystagmus, strabismus, and abnormalities of cranial nerve func-
likelihood that underlying immune deficiency is present. At the tion such as absent or decreased facial sensation, palpebral, men-
author’s hospital, disseminated protothecosis was diagnosed in ace, or gag reflexes.6,14,22-25 Dried fecal material or blood may
a canine renal transplant recipient that was treated with cyclo- be present around the anus, and rectal examination may reveal
sporine, and lymphocutaneous P. wickerhamii infection was the presence of hematochezia, increased fecal mucus, or melena.
diagnosed in a dog treated with chemotherapy for lymphoma.7
The median age of affected dogs in one series of 17 dogs was 4 Diagnosis
years (range, 18 months to 11 years), and approximately 70%
of the affected dogs were female.6 Cats with protothecosis have Laboratory Abnormalities
single or multiple nodular skin lesions; disseminated disease has Complete Blood Count and Serum Biochemical Tests
not been reported. Affected cats are typically FIV and FeLV The CBC in dogs with disseminated protothecosis is often nor-
negative, are otherwise in good health, and have ranged in age mal. Occasionally it reveals nonregenerative anemia and/or
from 3 to 16 years.8-13 leukocytosis due to a neutrophilia and sometimes monocyto-
sis.15,16,18 Mild bandemia and/or eosinophilia may be present in
Clinical Features some dogs. The serum biochemistry panel may also be normal
or reveal hyperglobulinemia due to a polyclonal gammopathy.
Signs and Their Pathogenesis Mild to moderate azotemia may be evident in dogs with renal
The pathogenesis of protothecosis is not fully understood. involvement.15,18 Hypoalbuminemia and hypocholesterolemia
Infection may occur secondary to cutaneous inoculation of may occur with severe bowel involvement.6
organisms, which may be followed by development of local-
ized cutaneous nodules or dissemination through lymphatic or Urinalysis
hematogenous spread. It is also possible that systemic invasion The urinalysis in dogs with disseminated protothecosis may be
by organisms that are ingested or colonize the gastrointestinal unremarkable or reveal isosthenuria, hematuria, pyuria, pro-
tract occurs. An incubation period of 10 days to several weeks teinuria, and the presence of Prototheca organisms in the sedi-
has been suggested for human protothecosis, based on recollec- ment.15 Urinalysis and urine culture should be performed in all
tion of penetrating injuries before onset of disease.2 dogs suspected to have protothecosis, because more than half of
In dogs, Prototheca can disseminate to a variety of tissues, affected dogs shed algae into the urine.
but especially the colon, eyes, brain and meninges, kidneys,
and long bones. This results in clinical signs of inappetence and Cerebrospinal Fluid Cytology
weight loss; acute or chronic large bowel diarrhea (often with CSF analysis in dogs with CNS protothecosis may be normal or
hematochezia and tenesmus); blindness; neurologic signs such reveal markedly increased total nucleated cell count (sometimes
as obtundation, seizures, or ataxia; polyuria and polydipsia; >1000 cells/µL) and increased CSF protein concentration (often
and sometimes lameness due to osteomyelitis.6,13-16 Deafness is >500 mg/dL).18,22,23 Eosinophilic pleocytosis has been described
often reported in association with disseminated protothecosis in some dogs,22,25 but eosinophils are not always present.23 Pro-
and may occur as a result of central nervous system (CNS) or totheca algae may also be found in the CSF.22,23
inner ear involvement.13,16,17 Ocular lesions include granuloma-
tous chorioretinitis, uveitis, and panophthalmitis. Other affected Diagnostic Imaging
organs include the liver, skeletal muscle, thyroid gland, lymph Plain Radiography
nodes, spleen, pancreas, stomach, small intestine, omentum, Thoracic radiographs in dogs with disseminated prototheco-
myocardium, aorta, spinal cord, and/or lungs.16-19 Involvement sis are usually unremarkable.15,18,19,21 Radiographs of affected
of these organs may lead to other signs such as vomiting, small long bones may reveal periosteal proliferative and/or osteolytic
bowel diarrhea, and/or melena. Sudden death has also been lesions due to osteomyelitis.6
reported, possibly secondary to myocardial involvement.6,20,21
Sonographic Findings
Physical Examination Findings Findings on abdominal ultrasound examination in dogs with dis-
Cutaneous lesions in dogs are nodular and may ulcerate. seminated protothecosis include hyperechogenicity of the renal
They often involve the footpads or distal limbs and associated cortices,15 a thickened colonic wall with loss of normal bowel
draining lymph nodes, but involvement of other sites such as wall layering,6 and abdominal lymphadenopathy. Some dogs
the trunk and planum nasale can also occur. In cats, single or have no abdominal sonographic abnormalities despite the pres-
multiple firm and sometimes ulcerated cutaneous nodules may ence of disseminated disease with colonic and renal involvement.7
be present. Lesions may be located on the head, footpads, and
distal limbs and tailbase8-10 and can resemble those caused by Other Imaging
Cryptococcus or Sporothrix. Colonoscopy in dogs with protothecosis that has disseminated
Dogs with disseminated disease may be obtunded and/or dehy- to the large intestine may reveal a hyperemic, friable, thickened,
drated and can have a thin body condition. Some dogs are febrile, irregular, and ulcerated mucosa.6
CHAPTER 70  Protothecosis 681

Magnetic resonance imaging of the brain of one dog with Cytologic Examination
CNS involvement revealed mild to moderate ventriculomegaly, Prototheca organisms may be seen using cytologic examina-
mild protrusion of the cerebellar vermis through the foramen tion of rectal scrapings; fine-needle aspirates of lymph nodes
magnum, syringomyelia, meningeal contrast enhancement, and or bone or skin lesions; CSF or vitreal or aqueous humor;
a focus of increased signal intensity in the brain on T2-weighted and urine sediment. The organisms are spheroid, ovoid, bean-
and fluid attenuated inversion recovery (FLAIR) sequences.23 shaped or elliptical, and range in diameter from 1.5 to 30 µm
As it enlarged, the focus developed contrast enhancement and (P. zopfii) or up to 10 µm (P. wickerhamii). They have a thick,
caused deformation of a lateral ventricle. hyaline cell wall that does not take up stain, and a basophilic,
granular cytoplasm. Multiple daughter cells (2 to usually
Microbiologic Tests <10) may be seen inside the cell (Figure 70-1). In the case of
Specific diagnostic assays for protothecosis are described in P. wickerhamii, the endospores are arranged symmetrically like
Table 70-2. a daisy or soccer ball, whereas the endospores of P. zopfii are

TABLE 70-2
Diagnostic Assays Available for Protothecosis
Assay Specimen Type Target Performance
Cytologic exami- Aspirates of affected tissues, Prototheca algae Usually large numbers of organisms are present, so sensi-
nation urine sediment, vitreous or tivity is high, but false negatives can still occur (such as
aqueous humor, CSF when urine sediment or CSF are examined). Organisms
may be confused with other fungi or Chlorella species,
so confirmation of the diagnosis requires culture.
Culture Urine, blood, CSF, vitreous or Prototheca Prototheca spp. grow rapidly (usually within 3 days) on
aqueous humor, aspirates or ­species routine media such as blood agar and Sabouraud dex-
biopsies of affected tissues trose agar. Allows species identification and subsequent
genotyping.
Histopathology Skin or colonic biopsies, Prototheca algae Usually large numbers of organisms are present, so
enucleated eyes, tissues col- sensitivity is high. Organisms are highlighted with PAS
lected at necropsy or silver stains. Organisms may be confused with other
fungi or Chlorella species, so confirmation of protothe-
cosis requires culture.

A B
FIGURE 70-1  A, Cytology of a subretinal aspirate from an 8-year-old intact male mixed breed dog with panophthalmitis due to Prototheca spp. infection. Moderate numbers of oval
to bean-shaped organisms are present in a proteinaceous background that contained variably degenerate and ruptured neutrophils. The organisms have a thin clear capsule with deep
purple cytoplasmic granulation. Sometimes endosporulation is evident (arrows). Wright’s stain, 1000× oil magnification. B, Cytology of a bone aspirate from a 2-year-old female mixed
breed dog that developed acute vision loss. Physical examination revealed bilateral retinal detachment and a nonpainful swelling of the right distal medial humerus. Abundant numbers of
Prototheca algae are present. (Courtesy Dr. Emily Pieczarka.)
682 SECTION 3  Fungal and Algal Diseases

distributed randomly.2 The algae may be mistaken for fungi identification to the species and genotype level in the clinical
such as Blastomyces, Coccidioides, or Cryptococcus. Because microbiology laboratory in the future.4,27
large numbers of organisms are typically present, the sensitiv- Antifungal susceptibility testing has been performed for Pro-
ity of cytologic examination for diagnosis of algal infection totheca but has not been standardized, and the clinical relevance
is high. However, failure to find organisms does not rule out of the results is not clear.2
protothecosis.
Molecular Diagnosis Using the Polymerase Chain Reaction
Culture Real-time PCR assays have been developed that detect and dif-
Prototheca species can be readily cultured in the microbiol- ferentiate Prototheca species,28 but these are not available on a
ogy laboratory on standard media (such as blood agar and commercial basis for diagnosis of protothecosis in animals.
Sabouraud dextrose agar), and yeast-like colonies are usually
visible within 7 days. Suitable specimens for culture include Pathologic Findings
aspirates or biopsies of affected tissue and body fluids such as Gross pathologic findings in dogs with disseminated protothe-
urine, CSF, and vitreous or aqueous humor. Prototheca zopfii cosis include enlarged lymph nodes and gray, tan, or white nod-
was also cultured from the blood of a dog with disseminated ular lesions or streaks in affected organs.13,16,18,25 The colonic
disease.21 Biochemical testing of isolates allows identification wall may be thickened and contain bloody fluid. Malacic foci
of the Prototheca species present and excludes other possible may be present in the brain, or the brain may appear normal
causes such as the green alga Chlorella, a very rare cause of dis- grossly, even when meningoencephalitis is present.7,24 Histo-
seminated disease in dogs that can closely resemble Prototheca pathologic examination of affected tissues typically reveals
in its microscopic appearance.2,26 Differentiation of P. zopfii large numbers of sporulating and nonsporulating algae accom-
genotypes requires PCR and ribosomal RNA gene sequenc- panied by a mixed inflammatory response and foci of necro-
ing. Matrix-assisted laser desorption/ionization–time of flight sis (Figure 70-2, A). In some tissues the inflammatory response
(MALDI-TOF) mass spectrometry offers promise for rapid can be minimal to absent.16,18 Organisms may be seen free and

A B

C
FIGURE 70-2  Histopathology of the renal lymph node of a 6-year-old spayed female shih tzu with protothecosis that was being treated with cyclosporine after renal transplanta-
tion. Numerous Prototheca organisms are present. The organisms range in diameter from 5 to 15 µm and have several morphologic forms. Sporulating forms are up to 15 µm in diameter
and contain two to four wedge-shaped endospores (daughter cells). Many of the organisms present as clear, refractile, oval to polyhedral bodies composed of a thick wall and devoid of
cytoplasm and nuclei. A, H&E stain. B, Periodic acid–Schiff stain. C, Gomori’s methenamine silver.
CHAPTER 70  Protothecosis 683

within macrophages. The cell wall stains well with Gomori’s Other treatments that may be required for protothecosis
methenamine silver and periodic acid–Schiff (PAS) stain, but in include intravenous fluid therapy, antiemetics, topical predniso-
contrast to Chlorella, cytoplasmic granules of Prototheca are lone acetate ophthalmic drops for uveitis, or ocular enucleation
PAS negative. Cutaneous lesions in cats have been associated to control pain and/or remove a focus of infection that fails to
with a granulomatous inflammatory response with multinucle- respond to chemotherapy (see Case Example). Other treatments
ated giant cells and, in some cases, necrosis and neutrophilic such as metronidazole, sulfasalazine, or dietary manipulation
inflammation.9,11 may help to control signs of protothecal colitis.

Treatment and Prognosis Public Health Aspects


Surgical excision can be curative for localized cutaneous Protothecosis is an uncommon disease of humans and most
protothecosis and should be performed whenever possible.9 often caused by P. wickerhamii. Infection is acquired from envi-
Medical treatment of protothecosis is challenging. In human ronmental sources, and direct transmission of other animals
patients, treatment is generally with amphotericin B, which or humans does not appear to be important. Mortality is low
in some cases results in complete cure. Addition of a tetra- (<5%), and two thirds of infections are chronic and localized
cycline has also been recommended.2 Outcomes with azole to the skin.2 Joint infections and tendonitis have been described
antifungals alone are often poor, although clinical responses after surgical or accidental wounds; some have been traced back
occur in some dogs with itraconazole alone.7 In one human to cleaning an aquarium.30 Disseminated infections and a syn-
patient, a combination of amikacin and tetracycline led to drome called olecranon bursitis (which follows elbow trauma)
cure.29 Treatment of canine disseminated protothecosis with account for the remaining third of cases. Underlying local or
amphotericin B leads to remission in some cases, but relapse systemic immune defects can be identified in many patients;
typically occurs when the drug is discontinued, or dogs fail topical, local, or systemic glucocorticoid treatment is the most
to respond to treatment and ultimately die or are eutha- frequently identified risk factor. Other immunosuppressive dis-
nized as a result of the disease.6,21 One dog with protothe- orders that may predispose humans to protothecosis include
cal meningoencephalitis appeared to improve clinically after diabetes mellitus and malignancy. Severe disseminated disease
treatment with oral itraconazole and intrathecal amphoteri- has been described in bone marrow and kidney transplant recip-
cin B, but was euthanized 4 weeks later because of progres- ients,31,32 in those with long-standing indwelling catheters or
sive disease.23 endotracheal tubes, and rarely in AIDS patients.30,33

CASE EXAMPLE Physical Examination:


Body Weight: 18.2 kg.
General: Bright, alert, responsive, and hydrated. T = 102.3°F
Signalment: “Merlot,” a 3-year-old female spayed Siberian (39.1°C), HR = 80 beats/min, panting, mucous membranes
husky dog from San Francisco, CA pink, CRT = 1 s.
History: Merlot was evaluated at the University of California, Integument, Eyes, Ears, Nose, and Throat: Dried barium was
Davis, Veterinary Medical Teaching Hospital for a 1-month present on the dog’s lips, and there were no other clinically
history of hemorrhagic diarrhea, inappetence, and significant findings. Fundoscopic examination revealed a
weight loss. Initially the diarrhea contained fresh blood region of retinal separation in the left ocular fundus that
and mucus but more recently contained only blood. The was adjacent to, dorsomedial to, and approximately the size
diarrhea occurred frequently and in small quantities. of the optic disc. The lesion appeared to contain yellowish
The owner reported that Merlot had lost approximately fluid.
5 kg over the past month and had increased thirst and Musculoskeletal: Thin but well-muscled. Body condition score
urination. There had been no history of vomiting, and was 3/9.
Merlot’s activity level was normal. Her diet consisted of a Cardiorespiratory: No clinically significant findings.
commercial dry dog food. Gastrointestinal/Genitourinary: No masses were palpated in
At a local veterinary clinic, multiple fecal flotations had been the abdomen. The dog vocalized and appeared painful on
negative, and Merlot had been treated with metronidazole, caudal abdominal palpation. Frank blood was present on
aminopentamide, bismuth subsalicylate, enrofloxacin, lop- the glove after rectal examination.
eramide, tetracycline, metoclopramide, and a highly digest- Peripheral Lymph Nodes: All peripheral lymph nodes were
ible diet without clinical response. A barium series had re- normal in size.
cently been performed, and findings were consistent with Laboratory Findings:
gastroenteritis. CBC:
Merlot had moved to San Francisco from Massachusetts HCT 48.6% (40%-55%)
4 months before the clinical signs began. On the way over MCV 70.9 fL (65-75 fL)
the owner stopped in Texas, where Merlot became lost in MCHC 34.6 g/dL (33-36 g/dL)
a field for 30 minutes. The owner reported that Merlot was WBC 12,100 cells/µL (6000-13,000 cells/µL)
taken to a dog park in San Francisco several times a week. No Neutrophils 9438 cells/µL (3000-10,500 cells/µL)
other history of illness existed. Lymphocytes 2057 cells/µL (1000-4000 cells/µL)
684 SECTION 3  Fungal and Algal Diseases

Monocytes 363 cells/µL (150-1200 cells/µL) to administer itraconazole (5 mg/kg PO q12h) and
Eosinophils 242 cells/µL (0-1500 cells/µL) lufenuron (5 mg/kg PO q24h). At a recheck examination
Platelets 259,000/µL (150,000-400,000 2 weeks later, the owners reported Merlot’s appetite had
platelets/µL). improved slightly and that she was defecating once to
Serum Chemistry Profile: twice a day. Each bowel movement was followed by a
Sodium 150 mmol/L (145-154 mmol/L) small amount of diarrhea at the end of defecation, with
Potassium 4.3 mmol/L (3.6-5.3 mmol/L) bloody mucoid material occasionally present. In the
Chloride 114 mmol/L (108-118 mmol/L) 2 weeks since her previous visit, a foxtail had been removed
Bicarbonate 28 mmol/L (16-26 mmol/L) from her right pelvic limb and she had been sprayed in
Phosphorus 4.2 mg/dL (3.0-6.2 mg/dL) the face by a skunk. Physical examination revealed a body
Calcium 9.8 mg/dL (9.7-11.5 mg/dl) weight of 19.2 kg (1 kg weight gain), but aqueous flare and
BUN 21 mg/dL (5-21 mg/dL) multifocal retinal separations were present in both eyes.
Creatinine 1.2 mg/dL (0.3-1.2 mg/dL) Urinalysis was unremarkable, and rectal scrapings showed no
Glucose 99 mg/dL (64-123 mg/dL) evidence of algal organisms. A biochemistry panel showed
Total protein 6.4 g/dL (5.4-7.6 g/dL) normalization of the creatinine and mildly increased activity
Albumin 2.9 g/dL (3.0-4.4 g/dL) of ALT, likely secondary to itraconazole administration. Topical
Globulin 3.5 g/dL (1.8-3.9 g/dL) prednisolone acetate ophthalmic drops and two additional
ALT 61 U/L (19-67 U/L) amphotericin B treatments were administered, but bilateral
AST 42 U/L (19-42 U/L) retinal detachment, blindness, then bilateral hyphema and
ALP 126 U/L (21-170 U/L) apparent ocular pain developed despite this treatment.
Cholesterol 145 mg/dL (135-361 mg/dL) Bilateral enucleation was performed. Histopathology of the
Total bilirubin 0 mg/dL (0-0.2 mg/dL). globes showed bilateral granulomatous uveitis and retinitis
Urinalysis: SGr 1.005; pH 6.0; no protein (SSA), bilirubin, with intralesional Prototheca organisms. In the left eye,
hemoprotein, or glucose; 0-1 WBC/HPF, rare RBC/HPF, rare posterior synechiae, intraocular hemorrhage, and cataract
transitional epithelial cells; no crystals, casts or bacteria. formation were identified.
Moderate numbers of encapsulated organisms were present At a recheck examination 3 months after enucleation, Merlot
that measured 10 × 17 µm. was eating well and had no signs of large bowel diarrhea.
Imaging Findings: Abdominal ultrasound examination: No Her body weight was stable and physical examination, a
abnormalities were identified. CBC, a biochemistry panel, and cytologic examination of a
Microbiologic Testing: Centrifugal zinc sulfate fecal rectal scraping were unremarkable. Itraconazole treatment
flotation: Negative, but yeast-like bodies were observed. was continued at a lower dose (5 mg/kg PO q24h), and the
Giardia and Cryptosporidium fluorescent antibody testing (fecal lufenuron was discontinued. Two months later, and shortly
specimen): Negative. after a new puppy was introduced, Merlot developed vomit-
Cytologic examination of rectal scrapings: Moderate numbers ing and diarrhea with small amounts of hematochezia, and
of neutrophils, small numbers of gram-positive coccobacilli, her body weight dropped to 17.5 kg. A serum biochemistry
and small numbers of what appeared to be algae were pres- panel showed mild hypoalbuminemia (2.6 g/dL). Cytologic
ent. examination of a rectal scraping again revealed Prototheca
Aerobic bacterial urine culture: Small numbers of Prototheca organisms, and a fecal flotation was negative. Merlot was
zopfii. treated with fenbendazole and the dose of itraconazole was
Algal susceptibility testing: MICs for amphotericin B, flucon- increased to 7.5 mg/kg PO daily, after which diarrhea resolved.
azole, and itraconazole were 0.5 µg/mL, >64 µg/mL, and 1 Four months later (14 months after initial diagnosis), the dog
µg/mL, respectively. remained in remission but was subsequently lost to follow-up.
Histopathology (rectal biopsy obtained by proctoscopy): ero- Comments: Diagnosis of protothecosis in this dog was
sive and hemorrhagic colitis, eosinophilic and lymphoplas- straightforward and initially based on routine urine sediment
macytic, moderate, with marked expansion of the lamina examination. Culture was then used to identify the causative
propria by intralesional spherical organisms (protothecal agent as P. zopfii. Colonic involvement was confirmed
colitis). using proctoscopy and biopsy. The most likely place that
Aerobic bacterial and fungal culture of a rectal biopsy: Small Prototheca infection was acquired was in Texas, but it is
numbers of mixed enteric species and P. zopfii. possible that infection was acquired elsewhere. Partial
Diagnosis: Disseminated P. zopfii infection with ocular and clinical remission was observed after treatment with lipid-
colonic involvement. complexed amphotericin B and itraconazole, but enucleation
Treatment: Merlot was initially treated with lipid-complexed was required to control pain. Remission was prolonged (>1
amphotericin B (3 mg/kg IV) on a Monday, Wednesday, year) even though only seven amphotericin B treatments
and Friday basis for five doses, after which increased were administered in the first 2 months after diagnosis.
serum creatinine concentration was noted (1.7 mg/dL). Lufenuron is a chitin synthetase inhibitor. Chitin is present in
She was also treated with oxytetracycline (21 mg/kg PO the cell walls of some algae (including Chlorella), but whether
q8h). She was discharged after 11 days with instructions Prototheca possess chitin in their cell walls is not clear.
CHAPTER 70  Protothecosis 685

SUGGESTED READINGS 17. Cook Jr JR, Tyler DE, Coulter DB, et al. Disseminated protothe-
cosis causing acute blindness and deafness in a dog. J Am Vet Med
Lass-Florl C, Mayr A. Human protothecosis. Clin Microbiol Rev. Assoc. 1984;184:1266-1272.
2007;20:230-242. 18. Rakich PM, Latimer KS. Altered immune function in a dog with dis-
Quigley RR, Knowles KE, Johnson GC. Disseminated chlorellosis in a seminated protothecosis. J Am Vet Med Assoc. 1984;185:681-683.
dog. Vet Pathol. 2009;46:439-443. 19. Buyukmihci N, Rubin LF, DePaoli A. Protothecosis with ocular
Stenner VJ, Mackay B, King T, et al. Protothecosis in 17 Australian dogs involvement in a dog. J Am Vet Med Assoc. 1975;167:158-161.
and a review of the canine literature. Med Mycol. 2007;45:249-266. 20. Blogg JR, Sykes JE. Sudden blindness associated with protothecosis
in a dog. Aust Vet J. 1995;72:147-149.
REFERENCES 21. Moore FM, Schmidt GM, Desai D, et  al. Unsuccessful treatment
1. Van Kruiningen HJ, Garner FM, Schiefer B. Protothecosis in a dog. of disseminated protothecosis in a dog. J Am Vet Med Assoc.
Pathol Vet. 1969;6:348-354. 1985;186:705-708.
2. Lass-Florl C, Mayr A. Human protothecosis. Clin Microbiol Rev. 22. Gupta A, Gumber S, Bauer RW, et  al. What is your diagnosis?
2007;20:230-242. Cerebrospinal fluid from a dog. Eosinophilic pleocytosis due to
3. Sonck CE, Koch Y. Prototheca as parasites of skin. Mykosen. protothecosis. Vet Clin Pathol. 2011;40:105-106.
1971;14:475-482. 23. Young M, Bush W, Sanchez M, et al. Serial MRI and CSF analysis
4. Ahrholdt J, Murugaiyan J, Straubinger RK, et al. Epidemiological in a dog treated with intrathecal amphotericin B for protothecosis.
analysis of worldwide bovine, canine and human clinical Proto- J Am Anim Hosp Assoc. 2012;48:125-131.
theca isolates by PCR genotyping and MALDI-TOF mass spec- 24. Salvadori C, Gandini G, Ballarini A, et  al. Protothecal granu-
trometry proteomic phenotyping. Med Mycol. 2012;50:234-243. lomatous meningoencephalitis in a dog. J Small Anim Pract.
5. Ricchi M, Goretti M, Branda E, et al. Molecular characterization 2008;49:531-535.
of Prototheca strains isolated from Italian dairy herds. J Dairy Sci. 25. Tyler DE, Lorenz MD, Blue JL, et al. Disseminated protothecosis
2010;93:4625-4631. with central nervous system involvement in a dog. J Am Vet Med
6. Stenner VJ, Mackay B, King T, et  al. Protothecosis in 17 Aus- Assoc. 1980;176:987-993.
tralian dogs and a review of the canine literature. Med Mycol. 26. Quigley RR, Knowles KE, Johnson GC. Disseminated chlorellosis
2007;45:249-266. in a dog. Vet Pathol. 2009;46:439-443.
7. Sykes JE. Unpublished observations, 2012. 27. von Bergen M, Eidner A, Schmidt F, et al. Identification of harm-
8. Coloe PJ, Allison JF. Protothecosis in a cat. J Am Vet Med Assoc. less and pathogenic algae of the genus Prototheca by MALDI-MS.
1982;180:78-79. Proteomics Clin Appl. 2009;3:774-784.
9. Dillberger JE, Homer B, Daubert D, et al. Protothecosis in two cats. 28. Ricchi M, Cammi G, Garbarino CA, et al. A rapid real-time PCR/
J Am Vet Med Assoc. 1988;192:1557-1559. DNA resolution melting method to identify Prototheca species.
10. Endo S, Sekiguchi M, Kishimoto Y, et  al. The first case of feline J Appl Microbiol. 2011;110:27-34.
Prototheca wickerhamii infection in Japan. J Vet Med Sci. 29. Zhao J, Liu W, Lv G, et al. Protothecosis successfully treated with
2010;72:1351-1353. amikacin combined with tetracyclines. Mycoses. 2004;47:156-158.
11. Finnie JW, Coloe PJ. Cutaneous protothecosis in a cat. Aust Vet J. 30. Pascual JS, Balos LL, Baer AN. Disseminated Prototheca wick-
1981;57:307-308. erhamii infection with arthritis and tenosynovitis. J Rheumatol.
12. Kaplan W, Chandler FW, Holzinger EA, et al. Protothecosis in a 2004;31:1861-1865.
cat: first recorded case. Sabouraudia. 1976;14:281-286. 31. Lass-Florl C, Fille M, Gunsilius E, et  al. Disseminated infection
13. Migaki G, Font RL, Sauer RM, et al. Canine protothecosis: review with Prototheca zopfii after unrelated stem cell transplantation for
of the literature and report of an additional case. J Am Vet Med leukemia. J Clin Microbiol. 2004;42:4907-4908.
Assoc. 1982;181:794-797. 32. Mohd Tap R, Sabaratnam P, Salleh MA, et  al. Characterization
14. Lane LV, Meinkoth JH, Brunker J, et  al. Disseminated protothe- of Prototheca wickerhamii isolated from disseminated algaemia
cosis diagnosed by evaluation of CSF in a dog. Vet Clin Pathol. of kidney transplant patient from Malaysia. Mycopathologia.
2012;41:147-152. 2012;173:173-178.
15. Pressler BM, Gookin JL, Sykes JE, et al. Urinary tract manifesta- 33. Kaminski ZC, Kapila R, Sharer LR, et  al. Meningitis due to
tions of protothecosis in dogs. J Vet Intern Med. 2005;19:115-119. Prototheca wickerhamii in a patient with AIDS. Clin Infect Dis.
16. Gaunt SD, McGrath RK, Cox HU. Disseminated protothecosis in a 1992;15:704-706.
dog. J Am Vet Med Assoc. 1984;185:906-907.
CHAPTER 71

Pneumocystosis
Remo Lobetti

it behaves similarly to a protozoan, because it is susceptible to


Overview of Pneumocystosis drugs used to treat protozoal infections but resistant to most
First Described: Pneumocystis was first discovered in Brazil antifungal drugs. The morphology of Pneumocystis organisms
in 1909 (Chagas), when it was initially mistaken for a try- and the histopathology of the lesions they produce in humans
panosome.1 It was first reported in dogs in the 1950s.2 and animals throughout the world are similar. Although con-
troversial, five species of Pneumocystis have been described,
Causes: Pneumocystis spp. (an ascomycete)
which appear to be highly host specific: two species that infect
Affected Hosts: Dogs, humans, pigs, horses, goats, nonhu- rats, P
­ neumocystis carinii and Pneumocystis wakefieldiae; one
man primates; Pneumocystis species appear to have host species that infects mice, Pneumocystis murina; a species that
specificity. infects rabbits, Pneumocystis oryctolagi; and P ­ neumocystis jir-
Geographic Distribution: Worldwide ovecii (pronounced “yee row vet zee”), which infects humans.5-7
Species designations for the organisms isolated from dogs
Mode of Transmission: Opportunistic invasion of immuno- and other animals require further genetic and biologic study,
suppressed hosts after transmission by aerosol although DNA analysis of Pneumocystis that infected a Cava-
Major Clinical Signs: Weight loss, respiratory distress, exercise lier King Charles spaniel suggested that dogs are infected with a
intolerance, variable cough unique species, with the proposed name of Pneumocystis canis.8
Differential Diagnoses: Canine distemper virus infection, Biologic differences between isolates from different hosts are
­bacterial pneumonia, mycobacterial infections, other supported by the relative difficulty of experimental cross-host
fungal causes of pneumonia, pulmonary neoplasia (espe- species transmission. Because historically P. carinii was thought
cially lymphoma), parasitic lung disease, acute respiratory to cause Pneumocystis pneumonia in humans, Pneumocystis
­distress syndrome, pulmonary fibrosis pneumonia has long been referred to as “PCP.” Despite the
name change to P. jirovecii in the 1970s, the term PCP has
Human Health Significance: Severely immunocompro- been retained for convenience. For purposes of this chapter, the
mised humans are at risk for pneumocystosis, but only organism will be referred to simply as Pneumocystis. Strain dif-
P. ­jirovecii infects humans. Whether humans can become ferences may also exist within species of Pneumocystis.
colonized with Pneumocystis strains that infect dogs is Pneumocystis appears to be maintained in nature by trans-
not known. mission from infected to susceptible hosts. The primary mode of
spread is thought to be airborne droplet transmission.9 Sporadic
disease may occur as a result of activation of latent infection by
Etiologic Agent and Epidemiology stress, crowding, and immunosuppressive therapy during hospi-
talization. Clinical disease can also be experimentally activated
Pneumocystis is a saprophyte of low virulence with a world- by treatment with glucocorticoids or cytotoxic chemotherapy. A
wide distribution. Its primary habitat is the mammalian lung, higher prevalence of infection was found in dogs with distemper
where it causes opportunistic pneumonia. Pneumocystis pneu- compared with a corresponding control population.10 Genetic
monia has been reported in dogs, pigs, horses, goats, primates, analysis of isolates from humans suggests that recent acquisition
and humans. Subclinical or latent infections are common in of infection from another person, rather than reactivation of
rats, mice, guinea pigs, rabbits, cats, sheep, and various wildlife infection, may be responsible for clinical illness.11,12
species. Most reports of pneumonia due to Pneumocystis spp. Most affected dogs have been young miniature dachshunds
infection are linked with documented or suspected immunodefi- (less than 1 year of age) or Cavalier King Charles spaniels.8,13-17
ciency syndromes in the host.3 Affected miniature dachshunds and Cavalier King Charles span-
The taxonomy of Pneumocystis is uncertain. Previously it iels may possess an underlying primary immune deficiency.17,18
was classified as a unicellular protozoan in the phylum Sar-
comastigophora. However, ultrastructurally the reproductive Clinical Features
behavior of Pneumocystis is similar to the ascospore formation
by yeasts, and its organelles and staining properties by light Signs and Their Pathogenesis
microscopy resemble those of pathogenic fungi. Phylogenetic Pneumocystis is inhaled and can colonize the lower respiratory
classification based on 16S-like rRNA sequences indicates that tracts of clinically healthy mammals; however, it rarely multiplies
Pneumocystis is most closely related to the fungi of the class to large numbers in the lungs of healthy hosts. Transmission of
Ascomycetes, especially Saccharomyces cerevisiae, and so it has infection to neonates can occur via aspiration of contaminated
been reclassified as fungus, “albeit an odd one.”4 Biologically, amniotic fluid when the placenta is infected. When impaired

686
CHAPTER 71  Pneumocystosis 687

host resistance (especially reduced CD4 ­T-lymphocyte counts) macrophages. With an adequate immune response, the body
or preexisting pulmonary disease is present, rapid proliferation eliminates the infection, but the removal of large numbers of
of organisms can occur.19 The entire life cycle of Pneumocystis is organisms and cellular debris may take up to 8 weeks. Both the
completed within alveolar spaces (Figure 71-1). In this location, presence of the organisms and the mild inflammatory response
two main forms, the trophozoite (1 to 4 µm in diameter) and they provoke contribute to pulmonary damage.
cyst (8 µm), are found. Trophozoites adhere closely to alveo- Although Pneumocystis infections are usually confined to
lar epithelial cells and are thought to undergo sexual reproduc- the lungs, organisms are rarely reported in extrapulmonary
tion by conjugation, with formation of a diploid zygote. The sites. In humans, extrapulmonary pneumocystosis occurs pri-
zygote becomes a sporocyte (or precyst) and undergoes meio- marily when there is overwhelming pulmonary infection, pro-
sis followed by mitosis. This leads to formation of a mature, found underlying immunodeficiency, and after long-term use
thick-walled cyst or spore case, which contains eight haploid of aerosolized pentamidine for prophylaxis against P. jirovecii
ascospores. The cyst ruptures, and the spores transform into tro- pneumonia in HIV-infected patients.20 Sites of extrapulmonary
phozoites, which replicate asexually to form more trophozoites, infection include lymph nodes, spleen, liver, bone marrow, gas-
or sexually to form more cyst forms. The transmissible form has trointestinal tract, eyes, thyroid gland, adrenal glands, kidneys,
not been identified.7 heart, pancreas, and external auditory canal. Extrapulmonary
The overgrowth and clustering of Pneumocystis within pneumocystosis has been described in one dog.
alveolar spaces leads to blockage of capillaries and decreased The typical clinical history in dogs with pneumocystosis is
gas exchange. Intra-alveolar organisms are often accompanied that of gradual weight loss and respiratory difficulty that pro-
by thickening of alveolar septa, but they seldom invade the pul- gresses over 1 to 4 weeks. The weight loss, which occurs in spite
monary interstitium and are rarely phagocytosed by alveolar of a good appetite in most dogs, may be associated with diar-
rhea and occasional vomiting. Cough is not always reported,
but reduced exercise tolerance is uniform. Infected animals
often show a minimal or temporary response to antibiotic and
Mitochondrion ­glucocorticoid therapy.

Physical Examination Findings


Nucleus
Abnormalities on physical examination of dogs with pneu-
mocystosis include dyspnea, tachycardia, and increased dry
respiratory sounds on thoracic auscultation. Animals are often
in poor condition and cachectic and can show dermatologic
changes, such as superficial bacterial pyoderma and demodico-
sis (Figure 71-2). Although the mucous membranes are gener-
ally of normal color, they may be cyanotic in severely affected
animals. Affected dogs remain relatively alert and afebrile,
A Asexual Haploid although mild pyrexia may be present.
trophic form

Conjugation
Excystment

Diploid
precyst

Meiosis

Early cyst

Maturation

B Sexual reproduction
FIGURE 71-1  Life cycle of Pneumocystis species. Trophozoites adhere to alveolar epi-
thelial cells in the lungs and undergo both asexual (A) and sexual (B) reproduction. Sexual
reproduction is initiated by conjugation, which results in formation of a zygote. Meiosis
occurs followed by mitosis, which leads to the formation of 8 ascospores within a mature
cyst. The cyst ruptures, releasing spores that become trophozoites. (Modified from Walzer
PD, Smulian AG. Pneumocystis species. In: Mandell GL, Bennett JE, Dolin R, eds. Principles FIGURE 71-2  One-year-old dachshund with pneumocystosis. Note the thin body
and Practice of Infectious Diseases, 7 ed. Philadelphia, PA: Elsevier; 2010:3377-3390.) condition and alopecia secondary to generalized demodectic mange.
688 SECTION 3  Fungal and Algal Diseases

FIGURE 71-3  Lateral thoracic radiographs from a 1-year-old male neutered Cavalier
King Charles spaniel with pneumocystosis. A diffuse marked interstitial pattern is present.
Mild right-sided cardiomegaly was also identified. FIGURE 71-4  Thoracic CT scan from a 4-year-old female spayed Cavalier King Charles
spaniel with pneumocystosis that was evaluated for lethargy, inappetence, and respira-
tory distress. There is a diffuse interstitial pattern and focal peripheral alveolar infiltrates
Diagnosis (arrowheads).

Laboratory Abnormalities
Complete Blood Count
Hematologic abnormalities in dogs with pneumocystosis are Microbiologic Tests
nonspecific. Neutrophilic leukocytosis is the most consistent Diagnostic assays available for pneumocystosis in dogs or cats
finding. Less frequently, eosinophilia or monocytosis are found. are described in Table 71-1. Identification of pneumonia in a
Lymphocyte counts can be elevated, normal, or low. Polycythe- Cavalier King Charles spaniel or miniature dachshund should
mia may occur secondary to arterial hypoxemia from impaired always raise suspicion for pneumocystosis.
gaseous exchange. Thrombocytosis is often present in miniature
dachshunds. Artifactual thrombocytopenia and megathrombo- Cytologic Diagnosis
cytosis have been observed in Cavalier King Charles spaniels, The diagnosis of pneumocystosis requires direct demonstration
which may have a hereditary basis that is likely unrelated to the of Pneumocystis cysts in biopsy specimens, respiratory fluids, or
suspected immunodeficiency. occasional extrapulmonary sites. Transtracheal, endotracheal,
or bronchoalveolar lavage specimens, lung aspirates, and oro-
Serum Chemistry Profile pharyngeal secretions may contain organisms. Transtracheal
Total serum proteins are usually within reference limits in dogs lavage specimens have been useful for detection of organisms
with pneumocystosis, with a low to borderline-low globulin in dogs.17 No cytologic technique is as reliable or as definitive
level, which correlates with low γ-globulin levels on serum pro- as histopathologic examination of lung biopsy specimens for
tein electrophoresis.17 In some dogs, mild hyperglobulinemia is documentation of active Pneumocystis infection. However, lung
present.21 Mild hypoalbuminemia may also be detected. Arte- biopsy is more invasive, has potential complications of hemor-
rial hypoxemia, hypocapnia, and increased arterial blood pH rhage or pneumothorax, and is associated with additional costs
indicate an uncompensated respiratory alkalosis. The arterial and hospitalization. Pneumocystis organisms are difficult to
pO2 is often lower than expected based on the clinical signs and detect in respiratory secretions or lavage specimens, and success
thoracic radiographs. in finding them usually depends on the experience of the exam-
iner and the collection and processing of specimens. Polychrome
Diagnostic Imaging stains, such as Wright’s, Giemsa, and methylene blue, will dem-
Findings on thoracic radiography include diffuse, bilaterally onstrate nuclei of trophozoites and intracystic sporozoites in
symmetric, miliary-interstitial to alveolar lung disease, with cytologic specimens, but the walls of cysts and trophozoites will
compensatory emphysema in severely infected animals.22 Inter- not be apparent (Figure 71-5). Diff Quik stain may be useful to
stitial patterns are most common (Figure 71-3). Solitary lesions, rapidly identify organisms.23
unilateral involvement, cavitary lesions, spontaneous pneumo-
thorax, and lobar infiltrates have also been described. Tracheal Serologic Diagnosis
elevation, right-sided heart enlargement, and pulmonary arte- Serologic assays are available for detection of human infections
rial enlargement reflect cor pulmonale secondary to diffuse pul- with P. jirovecii; however, their diagnostic value is uncertain,
monary disease. Computed tomography (CT) scans may show since many immunodeficient humans who develop pneu-
diffuse pulmonary interstitial infiltrates and small focal regions mocystosis fail to produce antibody titers, and healthy indi-
of increased opacity (Figure 71-4), but CT findings in dogs with viduals frequently have high titers. Increased antibody titers
pneumocystosis have not been extensively described. to P
­ neumocystis persist for long periods, offering a valuable
CHAPTER 71  Pneumocystosis 689

TABLE 71-1
Diagnostic Assays Available for Pneumocystosis in Dogs or Cats
Assay Specimen Type Target Performance
Cytologic examination Lung aspirates; transtracheal, Pneumocystis False negatives can occur when specimen size is small or
endotracheal, or bronchoal- trophozoites low numbers of organisms are present. Identification to
veolar lavage specimens and cysts the genus level requires immunostaining or PCR assay.
PCR assays Lung aspirates; transtracheal, Pneumocystis The sensitivity and specificity of assays may vary de-
endotracheal, or bronchoal- spp. DNA pending on assay design. Assays designed to detect
veolar lavage specimens P. jirovecii may not detect organisms that infect dogs.
In the absence of cytologic evidence of Pneumocystis
infection, the significance of a positive PCR result
must be interpreted in light of clinical findings, be-
cause of the potential for subclinical colonization.
Histopathology Lung biopsy or necropsy Pneumocystis Large numbers of organisms are usually found filling
specimens trophozoites alveolar spaces. Cyst forms are best demonstrated
and cysts with silver stains or periodic acid–Schiff. The identity
of the organisms can be confirmed with immunohisto-
chemistry or PCR assay.

Molecular Diagnosis Using the Polymerase Chain Reaction


PCR assays have been used to detect Pneumocystis in bron-
choalveolar lavage specimens from humans12,27-29 and in lung
tissue from dogs.16,30 The sensitivity of PCR assays on bron-
choalveolar lavage fluids from infected humans is greater than
that of conventional staining.31 DNA analysis of PCR products
has been used to type isolates from humans and animals.8 Some
PCR assays (especially specific real-time PCR assays) that detect
P. jirovecii may not detect Pneumocystis organisms that infect
dogs because of sequence differences.

Immunologic Findings
The majority of the immunologic studies of pneumocystosis
in dogs have been done in the miniature dachshund.17,32 Lym-
phocyte stimulation assay results, using phytohemagglutinin
and pokeweed mitogen, show severe immunosuppression,
especially when compared to controls, even though a normal
FIGURE 71-5  Impression smear of lung tissue from a 1-year-old male neutered Cava- lymphocyte count can be present. Immunoglobulin fraction
lier King Charles spaniel with pneumocystosis. Large foamy and activated macrophages quantification consistently reveals deficiencies of IgA, IgM, and
are present, as well as fewer neutrophils and lymphocytes. Abundant trophozoites are IgG. Low globulin levels, immunoglobulin deficiencies, and
present and range in size from 1 to 2 µm (arrowhead). Organisms can also be seen within decreased lymphocyte function have been reported in the Cava-
a cyst (arrow). Wright’s stain. lier King Charles spaniel.18,30 The absence of immunoglobulins
in the presence of pathologic changes is a significant finding,
since affected dogs have chronic infections, which should
index of infection in epidemiologic studies. However, they result in an immunoglobulin response. Immunoglobulin defi-
are of limited use for an immediate diagnosis.24 Circulating ciencies are still present after the Pneumocystis and skin infec-
Pneumocystis antigen has been detected in human serum by tions have resolved, which supports the presence of a primary
counter-immunoelectrophoresis and ELISA methods. How- defect. Lymphocyte transformation studies and the results of
ever, antigenemia also is found in up to 15% of clinically immunoglobulin quantification suggest that both T- and B-cell
healthy humans. Direct fluorescent antibody assays can specifi- abnormalities exist in affected dogs. Serum complement activ-
cally detect organisms in sputum, tracheal aspirates, or pulmo- ity in affected miniature dachshunds has been normal. CD3
nary tissue.25 Immunoperoxidase techniques also can be used and CD79a lymphocyte staining of lymph nodes and spleen
to identify Pneumocystis in impression smears or formalin- shows marked absence of B cells with the presence of T cells.
fixed, paraffin-embedded lung sections.26 Immunologic abnormalities in the miniature dachshund are
similar to those described in human patients that have common
Fungal Culture variable immunodeficiency syndrome (also known as acquired
Pneumocystis cannot be cultured in the laboratory from clinical or adult-onset hypogammaglobulinemia). Common variable
specimens. immunodeficiency syndrome is a primary immunodeficiency
690 SECTION 3  Fungal and Algal Diseases

disease characterized by little or no antibody production by B


lymphocytes, normal or decreased numbers of B lymphocytes,
and abnormal T-lymphocyte function.33

Pathologic Findings
Gross Pathologic Findings
Pathologic findings in pneumocystosis are primarily confined
to the lungs, although dissemination to regional lymph nodes,
spleen, liver, bone marrow, and other organs has been reported.
On gross examination, the lungs are firm, consolidated, and pale
brown or gray. They do not collapse when the chest cavity is
opened. Unlike many pneumonic processes, fluid is not expressed
from cut surfaces of the lung. The pulmonary and mediastinal
lymph nodes are often enlarged. Small amounts of fluid may be
found in the pleural cavity. Cardiac enlargement, when present,
has been right sided in all cases.

Histopathologic Findings FIGURE 71-6  Histopathology of the lung of an 18-month-old male neutered
On histologic examination, alveolar spaces are filled with Cavalier King Charles spaniel with pneumocystosis. Foamy, honeycomb-like material
aggregates of amorphous, foamy, eosinophilic material that fills the alveoli. Periodic acid–Schiff stain.
has a frothy, honeycomb-like pattern (Figure 71-6). A few
macrophages and detached alveolar lining cells may be pres- Treatment with TMS has been reported in 8 dogs;16,17,22 4
ent, but polymorphonuclear leukocytes are absent. Special were dachshunds that subsequently recovered.22 A TMS dose
stains are needed to identify cyst forms. Little or no phagocy- of 15 mg/kg, every 8 hours, or 30 mg/kg, every 12 hours, for 3
tosis of intact Pneumocystis organisms is present. However, weeks was used; the longest reported follow-up in these studies
nonviable organisms (such as those seen after treatment) are was for 4 months. Folic acid supplementation should be given
often phagocytosed, and macrophages may contain Gomori’s if adverse effects such as leukopenia and anemia are observed
methenamine silver (GMS)-positive granular material that or if long-term therapy is required. In human infections, resis-
represents the residuum of cyst wall degradation. In some tance to sulfonamides has been noted among some strains of
instances, alveolar septa are markedly thickened by dense P. jirovecii.34
accumulations of plasma cells, lymphocytes, and macrophages Other treatment options include atovaquone, clindamy-
(lymphoplasmacytic pneumonia). The septa may be widened cin and primaquine, and dapsone-trimethoprim combinations.
by fibrosis in chronic infections. With GMS stain, cyst forms Atovaquone is licensed for the treatment of human pneumo-
are spherical, ovoid, or crescent-shaped structures that range cystosis.35 This drug, which also has been used to treat canine
from 4 to 7 µm in diameter and have dot-like, argyrophilic babesiosis (see Chapter 10), is not as effective against Pneu-
(stain with silver), focal cyst wall thickenings. Cyst walls also mocystis infections in humans as pentamidine or TMS but has
can be demonstrated with other stains such as toluidine blue, lower toxicity. In humans, pentamidine or atovaquone has also
and they fluoresce when stained with orange G of a Papani- been administered by aerosol, but this is less effective than oral
colaou stain. The internal structure of trophozoites in tissue administration and is not recommended.36,37
sections and smears are best demonstrated with Giemsa stain. Combination therapy with clindamycin and primaquine is
On ultrastructural examination, intact alveoli are filled with now the preferred alternative to treatment with TMS in humans
compact aggregates of trophozoite and cyst forms. Trophozoites (e.g., for patients intolerant of TMS)37 but has not been reported
commonly line alveoli. for treatment of dogs. Dapsone and trimethoprim or pyrimeth-
Appropriate histologic staining is essential to ensure detec- amine, in combination, have been effective in experimental ani-
tion of Pneumocystis organisms. Routine hematoxylin and mals and in clinical trials in immunosuppressed humans with
eosin stain does not readily demonstrate the developmental pneumocystosis.38 Pneumocystis is resistant to azole antifungal
forms of Pneumocystis. Only the nuclei of intracystic sporo- drugs and amphotericin B (because it lacks ergosterol in its cell
zoites and trophozoites are stained. Various modifications of wall), but the anthelmintics benzimidazole and albendazole
methenamine silver staining can be employed to stain the cyst have been effective in experimental infections.39,40
walls brownish-black; trophozoites will not be detected. The Nonspecific immunostimulants such as cimetidine and
cyst wall also stains with periodic acid–Schiff stain (see Figure levamisole have been given adjunctively to treat affected min-
71-6). iature dachshunds,41 without much success. Supportive care is
essential for any patient with Pneumocystis pneumonia because
Treatment and Prognosis of disturbed alveolar gaseous exchange. Supplemental oxygen
therapy may be needed, and ventilatory assistance may also be
Specific antimicrobial chemotherapy is most beneficial when required. Immunosuppressive agents should be discontinued
pneumocystosis is suspected (usually based on the affected dog if possible. Antimicrobial chemotherapy of pulmonary pneu-
breed) or diagnosed during its early stages. The treatment of mocystosis results in a decline in arterial oxygen related to the
choice is trimethoprim and a sulfonamide (TMS). Pentamidine inflammatory reaction to dying organisms. Administration of
isethionate (an aromatic diamidine) has also been effective in anti-inflammatory doses of glucocorticoids during specific treat-
humans, but TMS is more effective and less toxic. TMS has also ment for P. jirovecii infection improves pulmonary function and
been used as prophylaxis in immunodeficient humans. survival in humans.37,42-44
CHAPTER 71  Pneumocystosis 691

Public Health Aspects but further studies are required to verify this. The primary
risk factor for clinical pneumocystosis in humans is immuno-
Pneumocystis organisms are ubiquitous in the environment. deficiency (especially HIV infection, hematologic malignancies
However, molecular and epidemiologic evidence suggests that and solid tumors, transplant recipients, and collagen vascular
spread may occur between humans because of localized out- disorders).37 Thus, the greatest risk for acquisition of infec-
breaks. Humans and animals are exposed to the same envi- tion from a pet would be if the animal was clinically ill from
ronmental sources of Pneumocystis. Pneumocystis species that Pneumocystis pneumonia and the person in close contact was
infect dogs are likely different from those that infect humans, immunocompromised.

CASE EXAMPLE Serum Chemistry Profile:


Sodium 147 mmol/L (145-154 mmol/L)
Potassium 4.6 mmol/L (3.6-5.3 mmol/L)
Signalment: “Tessa”, a 1-year-old intact female miniature Phosphorus 4.8 mg/dL (3.0-6.2 mg/dL)
dachshund (see Figure 71-2) Calcium 10.9 mg/dL (9.7-11.5 mg/dL)
History: Tessa was evaluated for a history of chronic tachypnea BUN 17 mg/dL (5-21 mg/dL)
and weight loss. The clinical signs had been nonresponsive Creatinine 0.5 mg/dL (0.3-1.2 mg/dL)
to various treatments that had included multiple Glucose 132 mg/dL (64-123 mg/dL)
antibiotics, bronchodilators, mucolytics, multivitamins, and Total protein 5.1 g/dL (5.3-7.5 g/dL)
glucocorticoids. From 2 months of age, she had a history of Albumin 3.5 g/dL (2.3-3.5 g/dL)
recurrent infections including otitis externa, hemorrhagic Globulin 1.6 g/dL (2-3.7 g/dL)
enteritis, tonsillitis, folliculitis, and pyogranulomatous ALT 65 U/L (10-60 U/L)
dermatitis. These conditions had responded each time to ALP 210 U/L (100-250 U/L)
supportive care. Cholesterol 196 mg/dL (135-361 mg/dL)
Physical Examination: Total bilirubin 0.1 mg/dL (0-0.2 mg/dL)
Body Weight: 4.5 kg. α-globulins 0.8 g/dL (0.8-1.6 g/dL)
General: Quiet, alert, and responsive. Ambulatory on all four β-globulins 0.6 g/dL (0.7-1.3 g/dL)
limbs. Rectal temperature = 101.8°F (38.8°C), HR = 138 γ-globulins 0.3 g/dL (0.4-1.1 g/dL).
beats/min, RR = 129 breaths/min, mucous membranes pink, Urinalysis: SGr 1.045; pH 6.0, negative for protein, bilirubin,
CRT = 1 s. blood, and glucose. Inactive sediment.
Integument: A poor coat was present with focal alopecia on Imaging Findings: On thoracic radiographs the
the head and back. cardiovascular structures appeared within normal limits.
Musculoskeletal: Body condition score was 3/9 with diffuse A diffuse interstitial pulmonary pattern was present.
and symmetrical muscle atrophy. Cytology Findings: Transtracheal wash cytology demon-
Respiratory: An increased respiratory rate with increased strated active, blastic alveolar macrophages with phagocyt-
abdominal effort was noted; tracheal palpation elicited a ic vacuolization, large numbers of neutrophils, and many
dry cough. free and phagocytosed Pneumocystis organisms.
All Other Systems: No clinically significant abnormalities were Aerobic Bacterial Culture: Culture of the transtracheal
noted. wash fluid yielded no growth after 48 hours’ incubation.
Laboratory Findings: Fecal Examination: Fecal flotation: Negative for parasites.
CBC: Diagnosis: Pneumocystis spp. pneumonia.
HCT 54% (40-55%) Treatment: The dog was treated with a potentiated
MCV 68 fL (65-75 fL) sulfonamide at 15 mg/kg PO q8h for 3 weeks, which resulted
MCHC 35.7 g/dL (33-36 g/dL) in a complete recovery.
WBC 34,600 cells/µL (6000-15,000 cells/µL) Comments: This dog was diagnosed with pneumocystosis as
Neutrophils 23,200 cells/µL (3000-11,000 cells/µL) a result of a primary immune deficiency disease, most likely
Band neutrophils 700 cells/µL (0-300 cells/µL) common variable immunodeficiency syndrome. The latter
Lymphocytes 5500 cells/µL (1000-4800 cells/µL) would also explain the history of chronic skin infections and
Monocytes 4800 cells/µL (100-2000 cells/µL) gastroenteritis. CD3 and CD79a lymphocyte staining of a
Basophils 350 cells/µL (100 cells/µL) peripheral lymph node biopsy showed a marked absence of
Platelets 730,000 platelets/µL (200,000-500,000 platelets/µL). B cells.

SUGGESTED READINGS REFERENCES


Chabe M, Aliouat-Denis CM, Delhaes L, et  al. Pneumocystis: from a 1. Miller R, Huang L. Pneumocystis jirovecii infection. Thorax.
doubtful unique entity to a group of highly diversified fungal species. 2004;59:731-733.
FEMS Yeast Res. 2011;11:2-17. 2. Sedlmeier H, Dahme E. Pneumocystis carinii infection in dog.
Watson PJ, Wotton P, Eastwood J, et al. Immunoglobulin deficiency in Zentralbl Allg Pathol. 1955;93:150-155.
Cavalier King Charles Spaniels with Pneumocystis pneumonia. J Vet 3. Lobetti R. Pneumocystis carinii infection in miniature dachshunds.
Intern Med. 2006;20:523-527. Compend Cont Educ Pract Vet. 2001;23:320-327.
692 SECTION 3  Fungal and Algal Diseases

4. Stringer JR. Pneumocystis. Int J Med Microbiol. 2002;292:391-404. 27. Kitada K, Oka S, Kimura S, et al. Detection of Pneumocystis cari-
5. Stringer JR, Beard CB, Miller RF, et  al. A new name (Pneumo- nii sequences by polymerase chain reaction: animal models and
cystis jiroveci) for Pneumocystis from humans. Emerg Infect Dis. clinical application to noninvasive specimens. J Clin Microbiol.
2002;8:891-896. 1991;29:1985-1990.
6. Hughes WT. Pneumocystis carinii vs. Pneumocystis jiroveci: 28. Lu JJ, Chen CH, Bartlett MS, et  al. Comparison of six different
another misnomer (response to Stringer et  al.). Emerg Infect Dis. PCR methods for detection of Pneumocystis carinii. J Clin Micro-
2003;9:276-277:author reply 277–279. biol. 1995;33:2785-2788.
7. Chabe M, Aliouat-Denis CM, Delhaes L, et al. Pneumocystis: from 29. Roux P, Lavrard I, Poirot JL, et al. Usefulness of PCR for detection
a doubtful unique entity to a group of highly diversified fungal spe- of Pneumocystis carinii DNA. J Clin Microbiol. 1994;32:2324-2326.
cies. FEMS Yeast Res. 2011;11:2-17. 30. Hagiwara Y, Fujiwara S, Takai H, et  al. Pneumocystis carinii
8. English K, Peters SE, Maskell DJ, et al. DNA analysis of Pneumo- pneumonia in a Cavalier King Charles Spaniel. J Vet Med Sci.
cystis infecting a Cavalier King Charles spaniel. J Eukaryot Micro- 2001;63:349-351.
biol. 2001(suppl):106S. 31. Flori P, Bellete B, Durand F, et al. Comparison between real-time
9. Bartlett MS, Smith JW. Pneumocystis carinii, an opportunist in immu- PCR, conventional PCR and different staining techniques for diag-
nocompromised patients. Clin Microbiol Rev. 1991;4:137-149. nosing Pneumocystis jiroveci pneumonia from bronchoalveolar
10. Sukura A, Laakkonen J, Rudback E. Occurrence of Pneumocystis lavage specimens. J Med Microbiol. 2004;53:603-607.
carinii in canine distemper. Acta Vet Scand. 1997;38:201-205. 32. Lobetti R. Common variable immunodeficiency in miniature
11. Beard CB, Carter JL, Keely SP, et  al. Genetic variation in Pneu- dachshunds affected with Pneumocystis carinii pneumonia. J Vet
mocystis carinii isolates from different geographic regions: implica- Diagn Invest. 2000;12:39-45.
tions for transmission. Emerg Infect Dis. 2000;6:265-272. 33. Cunningham-Rundles C, Knight AK. Common variable immune
12. Kovacs JA, Gill VJ, Meshnick S, et al. New insights into transmis- deficiency: reviews, continued puzzles, and a new registry. Immu-
sion, diagnosis, and drug treatment of Pneumocystis carinii pneu- nol Res. 2007;38:78-86.
monia. JAMA. 2001;286:2450-2460. 34. Calderon E, de la Horra C, Montes-Cano MA, et  al. [Genotypic
13. Brownlie SE. A retrospective study of diagnosis in 109 cases of canine resistance to sulfamide drugs among patients with Pneumocystis
lower respiratory disease. J Small Anim Pract. 1990;31:371-376. jiroveci pneumonia]. Med Clin (Barc). 2004;122:617-619.
14. Canfield PJ, Church DB, Malik R. Pneumocystis pneumonia in a 35. Stoeckle M, Tennenberg A. Atovaquone for Pneumocystis carinii
dog. Aust Vet Practit. 1993;23:150-154. pneumonia. Ann Intern Med. 1995;122:314:author reply 314–315.
15. Ramsey IK, Foster A, McKay J, et  al. Pneumocystis carinii 36. Chan C, Montaner J, Lefebvre EA, et al. Atovaquone suspension
pneumonia in two Cavalier King Charles spaniels. Vet Rec. compared with aerosolized pentamidine for prevention of Pneu-
1997;140:372-373. mocystis carinii pneumonia in human immunodeficiency virus–
16. Sukura A, Saari S, Jarvinen AK, et al. Pneumocystis carinii pneumonia infected subjects intolerant of trimethoprim or sulfonamides.
in dogs—a diagnostic challenge. J Vet Diagn Invest. 1996;8:124-130. J Infect Dis. 1999;180:369-376.
17. Lobetti RG, Leisewitz AL, Spencer JA. Pneumocystis carinii in the 37. Walzer PD, Smulian AG. Pneumocystis species. In: Mandell GL,
miniature dachshund: case report and literature review. J Small Bennett JE, Dolin R, eds. Principles and Practice of Infectious Dis-
Anim Pract. 1996;37:280-285. eases. 7th ed. Philadelphia, PA: Elsevier; 2010:3377-3390.
18. Watson PJ, Wotton P, Eastwood J, et  al. Immunoglobulin defi- 38. Hughes WT. Use of dapsone in the prevention and treatment
ciency in Cavalier King Charles Spaniels with Pneumocystis pneu- of Pneumocystis carinii pneumonia: a review. Clin Infect Dis.
monia. J Vet Intern Med. 2006;20:523-527. 1998;27:191-204.
19. Kelly MN, Shellito JE. Current understanding of Pneumocystis 39. Bartlett MS, Queener SF, Shaw MM, et al. Pneumocystis carinii is
immunology. Future Microbiol. 2010;5:43-65. resistant to imidazole antifungal agents. Antimicrob Agents Che-
20. Watts JC, Chandler FW. Evolving concepts of infection by Pneu- mother. 1994;38:1859-1861.
mocystis carinii. Pathol Annu. 1991(26 Pt 1):93-138. 40. Bartlett MS, Edlind TD, Lee CH, et al. Albendazole inhibits Pneu-
21. Sykes JE. Unpublished observations, 2012. mocystis carinii proliferation in inoculated immunosuppressed mice.
22. Kirberger RM, Lobetti RG. Radiographic aspects of Pneumocystis Antimicrob Agents Chemother. 1994;38:1834-1837.
carinii pneumonia in the miniature Dachshund. Vet Radiol Ultra- 41. Farrow BR, Watson AD, Hartley WJ, et al. Pneumocystis pneumo-
sound. 1998;39:313-317. nia in the dog. J Comp Pathol. 1972;82:447-453.
23. Cregan P, Yamamoto A, Lum A, et al. Comparison of four meth- 42. Consensus statement on the use of corticosteroids as adjunctive
ods for rapid detection of Pneumocystis carinii in respiratory speci- therapy for Pneumocystis pneumonia in the acquired immu-
mens. J Clin Microbiol. 1990;28:2432-2436. nodeficiency syndrome. The National Institutes of Health–
24. Daly KR, Huang L, Morris A, et al. Antibody response to Pneu- University of California Expert Panel for Corticosteroids as
mocystis jirovecii major surface glycoprotein. Emerg Infect Dis. Adjunctive Therapy for Pneumocystis Pneumonia. N Engl J Med.
2006;12:1231-1237. 1990;323:1500-1504.
25. Ng VL, Virani NA, Chaisson RE, et al. Rapid detection of Pneumo- 43. Bozzette SA, Finkelstein DM, Spector SA, et al. A randomized trial
cystis carinii using a direct fluorescent monoclonal antibody stain. of three anti-Pneumocystis agents in patients with advanced human
J Clin Microbiol. 1990;28:2228-2233. immunodeficiency virus infection. NIAID AIDS Clinical Trials
26. Kondo H, Hikita M, Ito M, et al. Immunohistochemical study of Pneu- Group N Engl J Med. 1995;332:693-699.
mocystis carinii infection in pigs: evaluation of Pneumocystis pneu- 44. Masur H. Prevention and treatment of Pneumocystis pneumonia.
monia and a retrospective investigation. Vet Rec. 2000;147:544-549. N Engl J Med. 1992;327:1853-1860.
SECTION 4
Protozoal Diseases

CHAPTER 72

Toxoplasmosis
Michael R. Lappin

gastrointestinal tract (also known as the enteroepithelial cycle)


Overview of Toxoplasmosis and pass environmentally resistant oocysts in feces (Figure
First Described: The organism that was ultimately named 72-1). Sporozoites develop within shed oocysts after 1 to 5 days
Toxoplasma gondii was first described in France in 1908 of exposure to oxygen and appropriate environmental tempera-
(Nicolle and Manceaux)1 ture and humidity (Figure 72-2). This process is known as sporu-
lation. After oocyst ingestion, released sporozoites can penetrate
Cause: Toxoplasma gondii, a coccidial protozoan parasite
the intestinal tracts of cats or intermediate hosts and disseminate
(phylum Apicomplexa)
in blood or lymph as tachyzoites during active infection. T. gon-
Affected Host Species: Cats are the definitive host and are dii can penetrate most mammalian cells and replicates asexually
the only species known to complete the sexual phase of within infected cells until the cell is destroyed. If an appropriate
T. gondii culminating in the passage of oocysts in feces. immune response occurs, replication of tachyzoites is attenuated
Cats and most other vertebrates can serve as intermedi- and slowly dividing bradyzoites develop that persist within cysts
ate hosts; invertebrates can serve as transport hosts by in extraintestinal tissues. These tissue cysts form readily in the
mechanical carriage of T. gondii oocysts. central nervous system (CNS), muscles, and visceral organs. Live
Geographic Distribution: T. gondii has a worldwide distribu- bradyzoites may persist in tissue cysts for the life of the host.
tion except in the absence of cats It is likely that most T. gondii–infected cats and dogs harbor
tissue cysts for life. Thus, the presence of serum antibodies is
Major Clinical Signs: Fever, ocular inflammation, ataxia, sei- likely to indicate current infection. T. gondii seroprevalence rates
zures, muscle pain, and respiratory distress are the most vary by the lifestyle of the cat or dog. In general, increased sero-
common clinical signs in cats; dogs have similar signs but prevalence correlates with increased age as a result of increased
develop illness less frequently than cats. risk of exposure over time. It also correlates with outdoor expo-
Differential Diagnoses: In the dog, Neospora caninum induces sure, since animals with access to the outdoors are most likely
the most similar clinical signs; many other chronic intra- to contact infected intermediate hosts. In a recent study of clini-
cellular bacterial infections and fungal infections in dogs cally ill cats, T. gondii antibodies were detected in 31.6% of the
and cats need to be considered a differential diagno- 12,628 cats tested.5 The seroprevalence was lowest in regions
ses for many of the clinical and laboratory abnormities with low humidity (Figure 72-3). In another study of feral cats in
induced by T. gondii. Florida, T. gondii antibodies were detected in 12.1% of the cats.6
Human Health Significance: There is significant risk to the Although T. gondii infection is common among cats, the oocyst
fetus after transplacental infection and to any immuno- shedding period is generally less than 21 days. Thus, detection of
compromised person. T. gondii oocysts in feline feces is uncommon. For example, in
two studies in the United States, T. gondii oocysts were detected
in feces of fewer than 1% of cats.7,8
Dogs do not produce T. gondii oocysts, but they can
Etiologic Agent and Epidemiology mechanically transmit oocysts after they ingest feline feces.9
The tissue phases of T. gondii infection occur in dogs and may
Toxoplasma gondii is a coccidian that is one of the most prev- be associated with clinical disease. Approximately 20% of dogs
alent parasites infecting warm-blooded vertebrates around in the United States are seropositive for T. gondii antibodies.10
the world.2-4 Only cats complete the sexual phase in the Before 1988, many dogs diagnosed with toxoplasmosis based

693
694 SECTION 4  Protozoal Diseases

FIGURE 72-1  Life cycle of Toxoplasma gondii. Cats are most often infected when they ingest bradyzoite cysts in tissues of prey. The bradyzoites transform into merozoites and undergo
repeated cycles of schizogony in the cat’s intestinal tract. The merozoites then transform into microgametes (male) and macrogametes (female). When a microgamete penetrates a mac-
rogamete, a zygote forms. The zygote is shed as an unsporulated oocyst in the feces, which is not infectious. Sporulation occurs after 1 to 5 days. The extraintestinal cycle occurs in cats
or other intermediate hosts after they ingest sporulated oocysts or tissue cysts. Sporozoites penetrate intestinal cells and transform into tachyzoites, which rapidly multiply in nucleated
cells throughout the body in the face of immunosuppression. This can result in severe disease in the developing fetus. Ultimately organisms are contained by the immune system within
bradyzoite cysts in muscle, brain, and visceral organs (latent infection).

15-19%
25-29%
30-34%
35-39%
40-45%
FIGURE 72-3  Continental United States regional seroprevalence in clinically ill cats.
Results were based on the presence of Toxoplasma gondii-specific IgG or IgM in serum.
Information was available from 3,640 serum specimens. (From Vollaire MR, Radecki SV,
Lappin MR. Seroprevalence of Toxoplasma gondii antibodies in clinically ill cats in the
FIGURE 72-2  Sporulated oocysts of Toxoplasma gondii. The oocysts are 10 × 12 µm United States. Am J Vet Res 2005;66:874-877.)
in diameter.
bradyzoites) or it can occur by transplacental transmission. Cats
on histologic evaluation were in fact infected with Neospora can also be infected through the transmammary route.11 In dogs,
caninum (see Chapter 73). evidence for venereal transmission also exists, and repeated
transplacental infection has been documented.12,13
Clinical Features Most cats are not coprophagic, so most are infected when
they ingest T. gondii bradyzoites in tissue cysts during carniv-
Signs and Their Pathogenesis orous feeding. After ingestion, the bradyzoites transform into
Infection of warm-blooded vertebrates occurs after ingestion of merozoites, which replicate in the epithelial cells of the intesti-
any of the three life stages of T. gondii (sporozoite, tachyzoites, nal tract, then transform into microgametes and macrogametes.
CHAPTER 72  Toxoplasmosis 695

When a microgamete penetrates a macrogamete, a zygote forms.


The zygote then transforms into an unsporulated oocyst, which
is shed in feces for 3 to 21 days. After sporulation (formation
of sporozoites within the oocyst), oocysts can survive in the
environment for months to years and are resistant to most dis-
infectants. The T. gondii oocyst shedding prepatent period is
stage dependent (ingestion of bradyzoites has a shorter prepa-
tent period than ingestion of sporozoites) but is not dose depen-
dent.14 In addition, transmission of T. gondii is most efficient
when cats consume tissue cysts (carnivorism) and when interme-
diate hosts consume oocysts (fecal-oral transmission). Infection
of rodents with T. gondii leads to clinical signs of altered behav-
ior, so that the rodent becomes less fearful of cats.15 This may
increase the likelihood that the definitive host (cat) will become
infected and potentiate the sexual phase of the organism.
Whether clinical toxoplasmosis develops is dependent on both
host and parasite effects. Some strains of T. gondii may be more
pathogenic than others, and some strains may have specific tissue
affinities, such as a tendency to cause ocular disease in cats.16,17
If a poor immune response is mounted after primary infection,
overwhelming tachyzoite replication that results in tissue necrosis
is the major cause of disease.2-4 This mechanism is also likely in FIGURE 72-4  Toxoplasma gondii–associated chorioretinitis in an experimentally
cats or dogs with chronic (latent) toxoplasmosis that then become inoculated cat.
immune suppressed. One example is activation of T. gondii
infection in cats or dogs after administration of immunosuppres- cause of uveitis in cats; anterior uveitis or posterior uveitis can
sive drugs such as cyclosporine.18-21 Other immunosuppressive occur, and the manifestations can be either unilateral or bilat-
conditions such as FIV infection can also result in activation of eral.2,41,42 Kittens infected transplacentally or through the trans-
toxoplasmosis.22 The mechanisms for chronic clinical toxoplas- mammary route commonly develop ocular disease.16
mosis have not been fully determined.2 T. gondii antigens and In dogs, respiratory, gastrointestinal, or neuromuscular infec-
antigen-containing immune complexes have been documented in tion with associated signs of fever, vomiting, diarrhea, respira-
the serum of affected cats. Persistent infection was not associated tory distress, ataxia, seizures, and icterus occurs most commonly
with chronic kidney failure in cats of one study.23-25 with generalized toxoplasmosis.2,43-50 Generalized toxoplasmo-
The large majority of cats infected with T. gondii never develop sis is most common in immunosuppressed dogs, such as those
detectable clinical abnormalities. In general, the enteroepithelial with canine distemper virus infection or those treated with cyclo-
cycle in the cat rarely leads to clinical signs. Only 10% to 20% of sporine to prevent renal transplant rejection. Neurologic signs
experimentally inoculated cats develop self-limiting, small bowel depend on the location of the primary lesions and include ataxia,
diarrhea for 1 to 2 weeks following primary oral inoculation with seizures, tremors, cranial nerve deficits, paresis, and paralysis.
T. gondii tissue cysts; this is presumed to be from the enteroepithe- Dogs with myositis present with weakness, stiff gait, or muscle
lial replication of the organism. T. gondii enteroepithelial stages atrophy. Rapid progression to tetraparesis and paralysis with
were found in intestinal tissues from two cats with inflammatory lower motor neuron dysfunction can occur. One study associated
bowel disease that had positive response to administration of anti- T. gondii antibodies with polyradiculoneuritis in dogs.49 Some
Toxoplasma drugs.26 Eosinophilic fibrosing gastritis was recently dogs with suspected neuromuscular toxoplasmosis probably have
described in a T. gondii–infected cat.27 neosporosis. Myocardial infection with ventricular arrhythmias
Fatal extraintestinal toxoplasmosis in cats can develop from occurs in some infected dogs. Retinitis, anterior uveitis, iridocycli-
overwhelming intracellular replication of tachyzoites following tis, nodular conjunctivitis, and optic neuritis occur in some dogs
primary infection; hepatic, pulmonary, CNS, and pancreatic tissues with toxoplasmosis, but they are less common than in cats.50
are commonly involved.2-4,28-30 Kittens infected by the transpla- Cutaneous disease also has been reported, which can manifest
cental or transmammary routes develop the most severe signs as pustules, pruritus, subcutaneous nodules, and/or alopecia.21,51
of extraintestinal toxoplasmosis and generally die of pulmonary
or hepatic disease. Common clinical signs in cats with dissemi- Diagnosis
nated toxoplasmosis include lethargy, anorexia, and respiratory
distress. Disseminated toxoplasmosis has been documented in Diagnosis of toxoplasmosis requires interpretation of specific
cats concurrently infected with FeLV, FIV, or feline infectious microbiologic tests (Table 72-1) in light of the presence of
peritonitis virus, as well as following cyclosporine administra- consistent clinical abnormalities. No clinical abnormalities are
tion for allergic dermatitis, immune-mediated disorders, or after specific for the disease.2-4
renal transplantation.18,19,31
Chronic toxoplasmosis occurs in some cats and should be Laboratory Abnormalities
on the differential diagnosis list for cats with uveitis, chorio- Complete Blood Count
retinitis (Figure 72-4), cutaneous lesions, fever, muscle hyper- T. gondii should be on the differential diagnosis list for cats
esthesia, myocarditis with arrhythmias, weight loss, anorexia, or dogs with appropriate clinical findings and nonregenerative
seizures, ataxia, icterus, diarrhea, respiratory distress, or pancre- anemia, neutrophilic leukocytosis or neutropenia, lymphocyto-
atitis.2-4,32-42 Toxoplasmosis appears to be a common infectious sis or lymphopenia, monocytosis, and/or eosinophilia.
696 SECTION 4  Protozoal Diseases

TABLE 72-1
Microbiologic Assays for Diagnosis of Toxoplasma gondii Infection
Assay Specimen Type Assay Target Comments
Fecal flotation Feces T. gondii oocysts The negative predictive value of this assay is poor for clinical
toxoplasmosis, since most cats cease shedding by the time
illness occurs.
Fecal PCR assay Feces T. gondii DNA As for fecal flotation.
Cytologic Effusions, skin T. gondii tachyzoites Confirms current infection and is generally associated with
­examination lesions, tissue aspi- and occasionally disease. Organisms cannot be readily distinguished from
rates bradyzoites Neospora caninum.
Histopathology Multiple tissues T. gondii tachyzoites If the organisms are detected in the presence of inflammation
and bradyzoites and necrosis, clinical toxoplasmosis is likely. Can be dif-
ficult to differentiate T. gondii from tissue protozoans such
as N. caninum in dogs. Immunohistochemistry can be used
to differentiate the two species.
PCR assay Effusions, blood, T. gondii DNA T. gondii DNA can be amplified from the blood of normal
multiple tissues dogs and cats and so PCR on blood has a low positive
predictive value. Amplification of specific DNA confirms
infection, and if appropriate clinical signs and inflammation
are present, positive results document clinical toxoplasmosis.
Serology for Serum T. gondii IgM Most consistent with recent infection but can be induced dur-
T. gondii IgM antibodies ing reinfection and by other immune drugs such as glucocor-
ticoids. Positive results do not correlate with active disease.*
T. gondii IgG Serum T. gondii IgG Most consistent with infection of > 10 days duration. Positive
antibodies results do not correlate with active disease.*
Agglutination Serum T. gondii Hypothetically detects all antibody classes but can be falsely
assays antibodies negative in animals that only possess anti-Toxoplasma
IgM antibody. Positive results do not correlate with active
disease.*

*Results of serum antibody assays are combined with clinical findings to aid in making a diagnosis of clinical toxoplasmosis.

Serum Chemistry Profile Sonography


Depending on the organ system involved, cats with clinical Ultrasound findings consistent with pancreatitis or diffuse hepa-
toxoplasmosis may have increases in serum protein and bili- titis could be noted in dogs or cats with involvement of these
rubin concentrations, as well as increased activities of creatine tissues due to T. gondii infection. Intra-abdominal lymphadeno-
kinase, ALT, ALP, and lipase.2-4 Findings are similar for dogs; megaly may be noted in dogs or cats with polysystemic disease.
dogs with chronic toxoplasmosis may develop hyperglobulin-
emia that is generally polyclonal.52 Advanced Imaging
MRI findings in dogs and cats with toxoplasmosis may be unre-
Urinalysis markable, or focal or mass lesions may be identified in the brain
Proteinuria and bilirubinuria have been detected in some dogs and/or spinal cord. On MRI, lesions are often isointense on
or cats with clinical toxoplasmosis. T1-weighted imaging and hyperintense on T2-weighted imag-
ing, and they may enhance peripherally or uniformly with con-
Cerebrospinal Fluid Analysis trast (Figure 72-6).53-55
CSF protein concentrations and cell counts are often higher
than normal, with the predominant white blood cells in CSF Microbiologic Testing
being small mononuclear cells. However, increased neutrophils Cytologic Diagnosis
also are commonly found in cats with acute CNS toxoplasmosis. Using cytologic examination, T. gondii bradyzoites or tachyzo-
Increased CSF protein concentrations and mixed inflammatory ites can be detected in tissues, effusions, bronchoalveolar lavage
cell infiltrates occur in dogs with CNS toxoplasmosis. fluids, aqueous humor, or CSF (Figure 72-7).32,35,36,38 A defini-
tive antemortem diagnosis of toxoplasmosis can be made if the
Diagnostic Imaging organism is identified; however, this is uncommon, particularly
Radiographs in association with sublethal disease. In the dog, N. caninum
Pulmonary toxoplasmosis most commonly causes diffuse inter- tachyzoites cannot be distinguished from those of T. gondii on
stitial to alveolar patterns (Figure 72-5); pleural effusion has cytologic examination, and so further diagnostics are needed to
rarely been documented as well. make a definitive diagnosis.2
CHAPTER 72  Toxoplasmosis 697

FIGURE 72-5  Thoracic radiographs of a 7-year-old female spayed domestic shorthair cat with pulmonic toxoplasmosis that developed one month after renal transplantation. There is
a diffuse interstitial pattern. The cat subsequently developed respiratory arrest and tachyzoites were seen in fluid that was suctioned from the endotracheal tube used for resuscitation. Nec-
ropsy revealed severe, diffuse, necrotizing pneumonia with intralesional tachyzoites. Histiocytic, lymphoplasmacytic, and necrotizing lesions with intralesional tachyzoites were also seen
in the renal allograft, ureter, bladder, liver, pancreas, peritoneum, muscularis mucosa of the stomach, and myocardium. Tachyzoites were not seen in the brain, but there was perivascular
inflammation that suggested the possibility of infection. Immunohistochemistry confirmed that the tachyzoites were T. gondii.

of oocysts in cats has a poor negative predictive value for clinical


toxoplasmosis, because most clinical toxoplasmosis results from
disseminated infections that occur after oocyst shedding has been
completed.

Serologic Diagnosis in Cats


Detection of T. gondii antibodies in serum is used most fre-
quently for the diagnosis of clinical toxoplasmosis. A multitude
of different techniques have been assessed, including ELISA,
immunofluorescent antibody (IFA), Western blot immunoassay,
and a variety of agglutination tests.57-67 A latex agglutination
assay and an indirect hemagglutination assay are available com-
mercially. These assays can be applied to serum from either dogs
or cats and theoretically detect all classes of immunoglobulin
directed against T. gondii. However, they rarely detect antibody
in feline serum samples when only IgM is present.60 ELISA, IFA,
and Western blot immunoassays have been adapted to detect
IgM, IgG, and IgA antibody responses by using heavy-chain–
specific secondary antibodies.57,63,64 T. gondii–specific serum
IgA antibody responses are similar to IgG antibody responses,
FIGURE 72-6  T1-weighted postcontrast sagittal magnetic resonance image from an and this antibody class is usually measured only in research
11-year-old female spayed domestic longhair that was evaluated for a 5-day history of studies. Several commercial laboratories in the United States
inappetence, ataxia, abnormal vocalization, and circling. The cat was FIV and FeLV negative offer T. gondii IgM and IgG testing by ELISA.68 The following
and had no other immunosuppressive conditions. A contrast-enhancing lesion is present are common findings concerning T. gondii IgM and IgG anti-
in the right brainstem. CSF analysis revealed a mixed monocytic (primarily lymphocytic) body test results in cats.
pleocytosis with a total nuclear cell count of 36 cells/µL and a total protein concentration of Toxoplasma gondii IgM antibody titers. 
78 mg/dL. Meningoencephalitis secondary to T. gondii infection was confirmed at necropsy. • Using ELISA, approximately 80% of healthy, experimen-
tally infected cats have detectable T. gondii–specific IgM in
Fecal Flotation serum within 2 to 4 weeks after inoculation with T. gon-
T. gondii oocysts measure 10 × 12 µm. When identified in feces dii; these titers generally are negative within 16 weeks after
of cats with diarrhea, this suggests infection by T. gondii.2-4,56 infection.57,58
However, Besnoitia and Hammondia infections of cats pro- • As occurs in some healthy women, persistent IgM titers (>16
duce morphologically similar oocysts. If oocysts of this size are weeks) have been documented commonly in cats co-infected
detected in feces of dogs, N. caninum infection is most likely if with FIV and in cats with ocular toxoplasmosis.41,65 Because
clinical disease is present.2 However, T. gondii oocysts can be of these findings and the fact that some cats never have a
present if the dog has ingested infected feline feces. PCR assays detectable IgM response, IgM titers cannot accurately be used
are available to differentiate T. gondii and N. caninum. Alter- to predict when a cat is shedding oocysts. If a clinician is con-
nately, serum antibody responses can be followed sequentially cerned that an individual cat is shedding T. gondii oocysts,
to attempt to determine the infecting genus. Overall, detection fecal flotation or a fecal PCR assay should be performed.
698 SECTION 4  Protozoal Diseases

A B
FIGURE 72-7  A, Tachyzoites of Toxoplasma gondii from the peritoneal cavity of a cat with fulminant toxoplasmosis. Unstained specimen. B, Cluster of tachyzoites in a tracheobronchial
lavage specimen from a 12-year-old male neutered cat that developed respiratory distress 3 weeks after renal transplantation. Wright’s stain, 1000× oil magnification.

• In one study of cats with clinical toxoplasmosis, T. gondii clinical toxoplasmosis will have reached their maximal IgG
IgM titers were detected in the serum of 93% of the cats, titer by the time they are evaluated serologically.
whereas T. gondii IgG titers were detected in only 60% of • Rising T. gondii IgG antibody titers occur in healthy infected
the cats. Thus, IgM antibodies have a higher positive predic- cats as well as cats with clinical toxoplasmosis and so, when
tive value than IgG for clinical feline toxoplasmosis.57,65 assessed alone, do not prove clinical toxoplasmosis.
• Some cats with chronic T. gondii infections that are initially • In humans and cats that reactivate chronic T. gondii infection
IgM positive, but then become IgM negative, again became after immune suppression, IgG titers only rarely increase.
IgM positive after repeat inoculation with T. gondii and In dogs, assays that detect IgM and IgG titers are available in some
immunosuppression through inoculation with FIV and commercial laboratories.68 There have been very few research
administration of glucocorticoids.57,67 However, clinical studies that assess canine serologic responses to T. gondii in differ-
signs of toxoplasmosis do not develop in these cats. Thus, ent situations. However, most of the findings documented in cats
presence of IgM antibodies in feline serum does not always appear to be true in dogs, based on the author’s clinical experience.
mean that clinical toxoplasmosis is present. For the reasons just discussed, antibody test results alone
Toxoplasma gondii IgG titers.  cannot be used to make a diagnosis of feline or canine toxo-
• T. gondii–specific IgG can be detected by ELISA in serum in plasmosis. However, the following combination can be used to
the majority of healthy experimentally inoculated cats within make a presumptive antemortem diagnosis:
3 to 4 weeks after infection.57,58 • Demonstration of antibodies in serum that suggest exposure
• By the time IgG antibodies are detected, the oocyst shedding to T. gondii
period has usually been completed. Thus, IgG-seropositive • Demonstration of an IgM titer higher than 1:64, or a four-
cats are of minimal public health risk. fold or greater rise in IgG titer, which suggests recent or
• T. gondii IgG antibody titers can be detected for at least active infection
6 years after infection in experimentally inoculated cats; • Clinical signs of disease consistent with toxoplasmosis
because the organism probably persists in tissues for life, IgG • Exclusion of other common causes of the clinical syndrome
antibodies probably do as well.62 • Positive response to appropriate treatment
• Single, high IgG titers do not necessarily suggest recent or Because T. gondii infection cannot be eliminated, most cats and
active T. gondii infection; healthy cats can have titers that dogs will be antibody positive for life, so there is little reason
exceed 1:10,000 as long as 6 years after experimental induction to repeat serum antibody titers after the clinical disease has
of toxoplasmosis.62 resolved or to administer drugs with T. gondii activity to cats or
• Some cats with low T. gondii antibody titers can become dogs without clinical signs of toxoplasmosis.
seronegative based on the cutoff value used in an individual Other serologic assays.  Assays that measure T. gondii anti-
assay even though T. gondii is still within tissues. Based on gens or immune complexes have been evaluated in some cats.
an approximate 10% interassay variation in the ELISA tech- However, as for antibody tests, antigen or immune complexes
nique, some cats with low positive IgG titers (1:64) can be can be detected in cats with or without clinical illness. These
positive for IgG antibodies on one analysis and negative on a assays are not offered commercially.23,24,57
subsequent analysis or vice versa.
• An increasing T. gondii IgG titer documents recent or active Molecular Diagnosis Using Nucleic Acid–Based Testing
infection, but in experimentally infected cats, the time span Recently, PCR assays have been used to document T. gondii DNA
from the first detectable positive IgG titer to the maximal IgG in feces and can be used to differentiate T. gondii from other
titer is approximately 2 to 3 weeks. Thus, some cats with organisms.56 T. gondii DNA can be amplified from the blood of
CHAPTER 72  Toxoplasmosis 699

A B C
FIGURE 72-8  Histopathologic findings in Toxoplasma gondii infections. A, A tissue cyst filled with Toxoplasma gondii bradyzoites in the brain tissues of an experimentally infected
mouse. B, Pancreas of the cat in Figure 72-5. There is pyogranulomatous inflammation with intralesional tachyzoites (arrows). H&E stain. C, Immunohistochemistry clearly demonstrates
the presence of T. gondii tachyzoites in the pancreas (brown staining).

TABLE 72-2
Antimicrobial Drugs That May Be Used to Treat Clinical Toxoplasmosis in Dogs and Cats
Drug Dose Route Interval Duration
Azithromycin 10 mg/kg PO q24h 4 weeks
Clindamycin 10-12 mg/kg PO q12h 4 weeks
Ponazuril 20 mg/kg PO q24h 4 weeks
Trimethoprim-sulfa 15 mg/kg PO q12h 4 weeks

If a positive response to treatment is achieved by week 4 but the animal is still improving slowly, continue treatment for 1 week past clinical resolu-
tion or when maximal response is recognized.

healthy cats and dogs, and so positive PCR assay results do not staining (Figure 72-8, C). Because T. gondii and N. caninum
correlate with clinical disease.69,70 Thus, PCR assays are used most induce similar syndromes in dogs, performance of PCR assays
frequently with cytologic examination or histopathology to docu- for both organisms in dogs is indicated in order to differentiate
ment that the organisms seen were T. gondii, or to detect T. gondii between them.
in aqueous humor or CSF in conjunction with serology.42,44-46,70
The combination of detection of T. gondii–specific antibody Treatment and Prognosis
in serum, blood, aqueous humor, or CSF and amplification of
T. gondii DNA by PCR assay is the most accurate way to diag- Cats or dogs with suspected clinical toxoplasmosis should be
nose ocular or CNS toxoplasmosis. Whereas T. gondii–specific administered supportive care as needed. Clindamycin hydro-
IgA, IgG, and DNA can be detected in aqueous humor and CSF chloride or a trimethoprim-sulfonamide combination has been
of both normal and clinically ill cats, T. gondii–specific IgM prescribed most frequently for the specific treatment of clinical
has only been detected in the aqueous humor or CSF of clini- toxoplasmosis (Table 72-2).2-4,66 One of the drugs should be
cally ill cats and so may be the best indicator of clinical disease. prescribed for 1 week, since most clinical signs of toxoplas-
Whether this is true for dogs has not been assessed to date. mosis will begin to resolve within that time period. If a positive
response is recognized, treatment should be continued for a
Pathologic Findings total of 4 weeks if possible. If there is a poor response to therapy
Gross Pathologic Findings after the first 7 days, an alternative drug should be considered.
T. gondii infection generally induces pyogranulomatous reac- Recurrence of clinical signs may be more common in cats or
tions and necrosis. The pyogranulomas may be visualized dogs treated for less than 4 weeks.72
grossly depending on the organ involved. Effusions that are Azithromycin has been used successfully in a limited num-
characteristic of modified transudates or exudates may be found ber of cats, but the optimal duration of therapy is unknown.
on necropsy. Pyrimethamine combined with sulfa drugs is effective for the
treatment of human toxoplasmosis but commonly results in
Histopathologic Findings toxicity in cats. Pyrimethamine has been administered suc-
Histopathology in animals with toxoplasmosis usually reveals cessfully with other drugs for the treatment of neosporosis in
pyogranulomatous inflammation with necrosis. T. gondii dogs and so may be effective for treatment of clinical toxo-
tachyzoites or bradyzoites are commonly detected (Figure 72-8, plasmosis in this species. Ponazuril has been used experimen-
A and B). However, if necrosis is severe, the organism may be tally in T. gondii–infected rodents and should be studied for
obscured. In these cases, DNA can often be amplified from the the treatment of feline toxoplasmosis.71,73 Currently, an optimal
tissues, or the agent can be visualized after immunohistochemical treatment regimen for the use of this drug for this purpose in
700 SECTION 4  Protozoal Diseases

cats is unknown. However, ponazuril (20 mg/kg PO q24h for


28 days) was apparently successful in the treatment of a dog
with suppurative keratitis and necrotizing conjunctivitis due
to T. gondii infection.50 Addition of fluconazole to pyrimeth-
amine and trimethoprim was also effective in rodent models
of toxoplasmosis and requires further study.74 Cats or dogs
with suspected Toxoplasma uveitis should be treated with anti-
Toxoplasma drugs in combination with topical or systemic glu-
cocorticoids to avoid secondary lens luxations and glaucoma.
Enucleation may be required if lens luxations and glaucoma
develop from persistent uveitis.
The prognosis is poor for cats or dogs with hepatic, CNS, or
pulmonary disease caused by tachyzoite replication, particularly
in those that are immunocompromised by anti-inflammatory
drugs or retrovirus co-infection. If they survive, cats or dogs
with CNS involvement may not experience complete resolution
of neurologic signs after treatment.

Immunity and Vaccination


FIGURE 72-9  Healed chorioretinitis in a 60-year-old woman who had recovered
There is no evidence to suggest that any drug can eliminate from congenital toxoplasmosis. The infection resulted in a permanent visual deficit. (Image
T. gondii infection in dogs or cats. Thus, recurrence of disease courtesy Mrs. Susan Sykes.)
is always possible, and infected dogs and cats often remain sero-
positive. In cats, if repeat oocyst shedding occurs, the number of studies, touching individual cats is probably not a common way
oocysts shed is small and shedding is of short duration. Systemic to acquire toxoplasmosis for the following reasons2-4,77-81:
reactivation of T. gondii infection after administration of immu- • Cats generally only shed oocysts for days to several weeks
nosuppressive drugs can occur in both dogs and cats. after primary inoculation.
Several types of vaccines have been studied to block oocyst • Repeat oocyst shedding is rare, even in cats receiving glucocor-
shedding in cats, but there is no vaccine for prevention of clinical ticoids or cyclosporine, or in those infected with FIV or FeLV.
toxoplasmosis in dogs or cats. • Cats inoculated with tissue cysts 16 months after primary
inoculation do not shed oocysts.
Prevention • Cats are very fastidious and usually do not allow feces to
remain on their skin for time periods long enough to lead to
To avoid exposure to T. gondii, cats and dogs should not be oocyst sporulation; the organism was not isolated from the
allowed to hunt or be fed undercooked meats. Care should be fur of cats that had been shedding millions of oocysts 7 days
taken to control transport hosts such as cockroaches that have previously.
been shown to carry T. gondii oocysts. However, because some cats will repeat oocyst shedding when
exposed a second time, cat feces should always be handled care-
Public Health Aspects fully. If a fecal specimen from a cat contains oocysts that mea-
sure 10 × 12 µm, it should be assumed that the organism present
Primary T. gondii infection in immunocompetent humans is T. gondii. The feces should be collected and removed daily
results in self-limiting fever, malaise, and lymphadenopathy that until the oocyst shedding period is complete; administration of
may not be recognized or is misdiagnosed. Primary infection of clindamycin (20 mg/kg PO daily) may shorten the oocyst shed-
mothers by T. gondii during gestation can lead to clinical toxo- ding period if started after infection is documented.2,82
plasmosis in the fetus; stillbirth, CNS disease, and ocular disease Because humans are not commonly infected with T. gondii
are common clinical manifestations (Figure 72-9). As T-helper through contact with individual cats, testing healthy cats for
cell counts decline, approximately 10% of people with AIDS evidence of T. gondii infection is not recommended.79 Fecal
develop toxoplasmic encephalitis from reactivation of bradyzo- examination is an adequate procedure to determine when cats
ites in tissue cysts. are actively shedding oocysts but cannot indicate when a cat has
People most commonly acquire toxoplasmosis by ingestion shed oocysts in the past. There is no serologic assay that accu-
of sporulated oocysts or tissue cysts, or transplacentally. To rately indicates this, and most cats that are shedding oocysts are
prevent toxoplasmosis, humans should avoid consumption of seronegative. Most seropositive cats have completed the oocyst
undercooked meats or ingestion of sporulated oocysts. In a shedding period and are unlikely to repeat shedding; most sero-
study of 6282 meat samples from 698 retail meat stores, using negative cats would shed the organism if infected. If owners are
a feline bioassay, T. gondii was detected in none of the beef concerned that they may have toxoplasmosis, they should see
or chicken specimens tested and only a small number of pork their physicians for testing.
specimens.75 However, T. gondii has been detected in the tissues Dogs do not complete the enteroepithelial phase of T. ­gondii
of free-ranging chickens.76 Although exposure to cats is epide- but can mechanically transmit oocysts after ingesting feline
miologically associated with acquiring toxoplasmosis in some feces.9
CHAPTER 72  Toxoplasmosis 701

CASE EXAMPLE Laboratory Findings:


Complete Blood Count: All values were within reference
ranges.
Signalment: “Fluffy,” a 3-year-old male neutered Siamese mix Serum Biochemistry Profile: All values were within reference
from Evans in northern Colorado ranges except for the activity of serum AST (190 U/L;
History: Fluffy was referred after being treated for anterior reference range, 19-42 U/L) and the activity of serum CK
uveitis and recurrent fever at another clinic. The clinical (2000 U/L; reference range, 50-125 U/L).
signs had been noted for approximately 1 month. Fluffy Urinalysis: No significant abnormalities; SGr = 1.041.
was entirely indoors in a suburban environment, but Imaging Findings: Thoracic and abdominal radiographs and
adopted from a humane society at around 6 months of abdominal ultrasound were offered but declined by the
age. His last vaccines (rabies, feline herpesvirus-1, feline owner.
calicivirus, and panleukopenia virus) were administered Microbiologic Testing: Serologic testing: FeLV antigen
6 months before the uveitis was first noted. He was fed a negative; FIV antibody negative; serum Bartonella IgG
commercial processed food. There were no other animals negative; Bartonella PCR assay negative on whole blood; T.
at the house. He was treated with selamectin monthly but gondii serum IgM 1:256; T. gondii serum IgG 1:1024.
had not received his dose this month because of the fever. Diagnosis: Suspected chronic, and possibly recurrent,
The fever seemed intermittent, and when normal, Fluffy toxoplasmosis.
had normal appetite and attitude. The uveitis persisted Treatment: Clindamycin (12 mg/kg PO q12h) was administered
in spite of treatment with amoxicillin-clavulanic acid for 28 days; fever resolved on day 3 of treatment. Prednisolone
administered daily for 10 days and topical 1% prednisolone acetate was continued every 6 hours topically bilaterally for
acetate applied three times daily. the first 14 days and was discontinued after anterior uveitis
Physical Examination: resolved. Subsequently, chorioretinal scars were noted on
Body Weight: 4.2 kg fundoscopic examination.
General: Quiet, alert, and responsive. Ambulatory on all four Comments: The findings were suggestive of chronic recurrent
limbs. T = 104.3°F (40.2°C), HR = 180 beats/min, RR = 30 toxoplasmosis, which does not respond to treatment with
breaths/min, mucous membranes pink, CRT = 1 s. β-lactam antibiotics. Myositis or pancreatitis may have
Integument: A dull haircoat was present. There was no contributed to some of the clinical and laboratory findings.
evidence of ectoparasites. These tissues commonly contain tissue cysts with T. gondii
Eyes: Bilateral anterior uveitis was present; fundoscopic bradyzoites that can repeatedly replicate as tachyzoites,
examination was difficult due to aqueous flare. which in turn results in acute disease. T. gondii IgM can be
Musculoskeletal: Body condition score was 4/9; possible detected in some cats with recurrent disease. Fluffy may have
discomfort was detected on muscle palpation. been infected with T. gondii when he was a stray and had an
Gastrointestinal: The cranial abdomen was tense with exacerbation or alternatively may have been infected more
abdominal splinting noted on palpation. recently in the home environment by ingestion of a transport
All Other Systems: No significant abnormalities were noted. host (cockroach, etc.) or intermediate host (rodent).

SUGGESTED READINGS 6. Luria BJ, Levy JK, Lappin MR, et  al. Prevalence of infectious
diseases in feral cats in Northern Florida. J Feline Med Surg.
Jones JL, Dubey JP. Foodborne toxoplasmosis. Clin Infect Dis. 2004;6:287-296.
2012;55:845-851. 7. Spain CV, Scarlett JM, Wade SE, et al. Prevalence of enteric zoo-
Robert-Gangneux F, Dardé M-L. Epidemiology and diagnostic strate- notic agents in cats less than 1 year old in central New York State.
gies for toxoplasmosis. Clin Microbiol Rev. 2012;25:264-296. J Vet Intern Med. 2001;15:33-38.
8. Dabritz HA, Miller MA, Atwill ER. Detection of Toxoplasma
REFERENCES gondii–like oocysts in cat feces and estimates of the environmental
1. Nicolle C, Manceaux L. Sur une infection à corps de Leish- oocyst burden. J Am Vet Med Assoc. 2007;231:1676-1684.
man (ou organismes voisins) du gondii. C R Seances Acad Sci. 9. Lindsay DS, et al. Mechanical transmission of Toxoplasma gondii
1908;147:763-766. oocysts by dogs. Vet Parasitol. 1997;73:27-33.
2. Dubey JP, Lindsay DS, Lappin MR. Toxoplasmosis and other 10. Levy JK, Lappin MR, Glaser AL, et al. Prevalence of infectious dis-
intestinal coccidial infections in cats and dogs. Vet Clin North Am eases in cats and dogs rescued following Hurricane Katrina. J Am Vet
Small Anim Pract. 2009;39:1009-1034. Med Assoc. 2011;238:311-317.
3. Lappin MR. Update on the diagnosis and management of Toxo- 11. Powell CC, Brewer M, Lappin MR. Detection of Toxoplasma gon-
plasma gondii infection in cats. Top Companion Anim Med. dii in the milk of experimentally infected lactating cats. J Vet Para-
2010;25:136-141. sitol. 2001;102:29-33.
4. Lappin MR. Toxoplasmosis. In: Couto G, Nelson R, eds. Small 12. Arantes TP, Lopes WD, Ferreira RM, et  al. Toxoplasma gondii:
Animal Internal Medicine. St Louis, MO: Mosby Elsevier; evidence for the transmission by semen in dogs. Exp Parasitol.
2010:1366-1373. 2009;123:190-194.
5. Vollaire MR, Radecki SV, Lappin MR. Seroprevalence of Toxo- 13. Bresciani KD, Costa AJ, Toniollo GH, et al. Transplacental trans-
plasma gondii antibodies in clinically ill cats in the United States. mission of Toxoplasma gondii in reinfected pregnant female
Am J Vet Res. 2005;66:874-877. canines. Parasitol Res. 2009;104:1213-1217.
702 SECTION 4  Protozoal Diseases

14. Dubey JP. Comparative infectivity of oocysts and bradyzoites of 38. Falzone C, Baroni M, De Lorenzi D, et al. Toxoplasma gondii brain
Toxoplasma gondii for intermediate (mice) and definitive (cats) granuloma in a cat: diagnosis using cytology from an intraoperative
hosts. Vet Parasitol. 2006;140:69-75. sample and sequential magnetic resonance imaging. J Small Anim
15. Vyas A, Kim SK, Giacomini N, et al. Behavioral changes induced Pract. 2008;49:95-99.
by Toxoplasma infection of rodents are highly specific to aversion 39. Kul O, Atmaca HT, Deniz A, et al. Clinicopathologic diagnosis of
of cat odors. Proc Natl Acad Sci U S A. 2007;104:6442-6447. cutaneous toxoplasmosis in an Angora cat. Berl Munch Tierarztl
16. Powell CC, Lappin MR. Clinical ocular toxoplasmosis in neonatal Wochenschr. 2011;124:386-389.
kittens. Vet Ophthalmol. 2001;4:87-92. 40. Lindsay SA, Barrs VR, Child G, et al. Myelitis due to reactivated
17. Spycher A, Geigy C, Howard J, et  al. Isolation and genotyp- spinal toxoplasmosis in a cat. J Feline Med Surg. 2010;12:818-821.
ing of Toxoplasma gondii causing fatal systemic toxoplasmo- 41. Lappin MR, Roberts SM, Davidson MG, et  al. Enzyme-linked
sis in an immunocompetent 10-year-old cat. J Vet Diagn Invest. immunosorbent assays for the detection of Toxoplasma gondii–
2011;23:104-108. specific antibodies and antigens in the aqueous humor of cats. J Am
18. Beatty J, Barrs V. Acute toxoplasmosis in two cats on cyclosporin Vet Med Assoc. 1992;201:1010-1016.
therapy. Aust Vet J. 2003;81:339. 42. Powell CC, McInnis CL, Fontenelle JP, et  al. Bartonella spe-
19. Bernsteen L, Gregory CR, Aronson LR, et al. Acute toxoplasmosis cies, feline herpesvirus-1, and Toxoplasma gondii PCR assay
following renal transplantation in three cats and a dog. J Am Vet results from blood and aqueous humor samples from 104 cats
Med Assoc. 1999;215:1123-1126. with naturally occurring endogenous uveitis. J Feline Med Surg.
20. Lappin MR, Scorza V. Toxoplasma gondii oocyst shedding in nor- 2010;12:923-938.
mal cats and cats treated with cyclosporine [Abstract]. Proceedings 43. Dubey JP, Carpenter JL, Topper MJ, et al. Fatal toxoplasmosis in
of the American College of Veterinary Internal Medicine Annual dogs. J Am Anim Hosp Assoc. 1989;25:659-664.
Forum, Denver CO, June 16, 2011. 44. Al-Qassab S, Reichel MP, Su C, et  al. Isolation of Toxoplasma
21. Webb JA, et  al. Cutaneous manifestations of disseminated toxo- gondii from the brain of a dog in Australia and its biological and
plasmosis in an immunosuppressed dog. J Am Anim Hosp Assoc. molecular characterization. Vet Parasitol. 2009;164:335-339.
2005;41:198-202. 45. da Silva AV, Pezerico SB, de Lima VY, et al. Genotyping of Toxo-
22. Davidson MG, Rottman JB, English RV, et al. Feline immunode- plasma gondii strains isolated from dogs with neurological signs.
ficiency virus predisposes cats to acute generalized toxoplasmosis. Vet Parasitol. 2005;127:23-27.
Am J Pathol. 1993;143:1486-1497. 46. Langoni H, Matteucci G, Medici B, et  al. Detection and molec-
23. Lappin MR, Cayatte S, Powell CC, et al. Detection of Toxoplasma ular analysis of Toxoplasma gondii and Neospora caninum
gondii antigen containing immune complexes in the serum of cats. from dogs with neurological disorders. Rev Soc Bras Med Trop.
Am J Vet Res. 1993;54:415-419. 2012;45:365-368.
24. Lappin MR, Greene CE, Prestwood AK, et  al. Enzyme-linked 47. Plugge NF, Ferreira FM, Richartz RR, et  al. Occurrence of
immunosorbent assay for the detection of circulating anti- antibodies against Neospora caninum and/or Toxoplasma gon-
gens of Toxoplasma gondii in the serum of cats. Am J Vet Res. dii in dogs with neurological signs. Rev Bras Parasitol Vet.
1989;50:1586-1590. 2011;20:202-206.
25. Hsu V, Grant DC, Zajac AM, et al. Prevalence of IgG antibodies 48. Dubey JP, Chapman JL, Rosenthal BM, et  al. Clinical Sarcocys-
to Encephalitozoon cuniculi and Toxoplasma gondii in cats with tis neurona, Sarcocystis canis, Toxoplasma gondii, and Neospora
and without chronic kidney disease from Virginia. Vet Parasitol. caninum infections in dogs. Vet Parasitol. 2006;137:36-49.
2011;176:23-26. 49. Holt N, Murray M, Cuddon PA, et al. Seroprevalence of various
26. Peterson JL, Willard MD, Lees GE, et  al. Toxoplasmosis in two infectious agents in dogs with suspected acute canine polyradiculo-
cats with inflammatory intestinal disease. J Am Vet Med Assoc. neuritis. J Vet Intern Med. 2011;25:261-266.
1991;99:473-476. 50. Swinger RL, Schmidt Jr KA, Dubielzig RR. Keratoconjunctivitis
27. McConnell JF, Sparkes AH, Blunden AS, et  al. Eosinophilic associated with Toxoplasma gondii in a dog. Vet Ophthalmol.
fibrosing gastritis and toxoplasmosis in a cat. J Feline Med Surg. 2009;12:56-60.
2007;9:82-86. 51. Hoffmann AR, Cadieu J, Kiupel M, et al. Cutaneous toxoplasmosis
28. Dubey JP, Carpenter JL. Histologically confirmed clinical toxo- in two dogs. J Vet Diagn Invest. 2012;24:636-640.
plasmosis in cats: 100 cases (1952-1990). J Am Vet Med Assoc. 52. Yarim GF, Nisbet C, Oncel T, et al. Serum protein alterations in
1993;203:1556-1566. dogs naturally infected with Toxoplasma gondii. Parasitol Res.
29. Dubey JP, Johnstone I. Fatal neonatal toxoplasmosis in cats. J Am 2007;101:1197-1202.
Anim Hosp Assoc. 1982;18:461-467. 53. Pfohl JC, Dewey CW. Intracranial Toxoplasma gondii granuloma
30. Dubey JP, Carpenter JL. Neonatal toxoplasmosis in littermate cats. in a cat. J Feline Med Surg. 2005;7(6):369-374.
J Am Vet Med Assoc. 1993;203:1546-1549. 54. Falzone C, Baroni M, De Lorenzi D, et al. Toxoplasma gondii brain
31. Nordquist BC, Aronson LR. Pyogranulomatous cystitis associated granuloma in a cat: diagnosis using cytology from an intraoperative
with Toxoplasma gondii infection in a cat after renal transplanta- sample and sequential magnetic resonance imaging. J Small Anim
tion. J Am Vet Med Assoc. 2008;232:1010-1012. Pract. 2008;49(2):95-99.
32. Park CH, Ikadai H, Yoshida E, et al. Cutaneous toxoplasmosis in a 55. Alves L, Gorgas D, Vandevelde M, et  al. Segmental meningomy-
female Japanese cat. Vet Pathol. 2007;44:683-687. elitis in 2 cats caused by Toxoplasma gondii. J Vet Intern Med.
33. Pfohl JC, Dewey CW. Intracranial Toxoplasma gondii granuloma 2011;25:148-152.
in a cat. J Feline Med Surg. 2005;7:369-374. 56. Salant H, Spira DT, Hamburger J. A comparative analysis of
34. Simpson KE, Devine BC, Gunn-Moore D. Suspected Toxoplasma- coprologic diagnostic methods for detection of Toxoplasma gondii
associated myocarditis in a cat. J Feline Med Surg. 2005;7:203-208. in cats. Am J Trop Med Hyg. 2010;82:865-870.
35. Hawkins EC, Davidson MG, Meuten DJ, et al. Cytologic identifica- 57. Lappin MR. Feline toxoplasmosis: interpretation of diagnostic test
tion of Toxoplasma gondii in bronchoalveolar lavage fluid of exper- results. Semin Vet Med Surg. 1996;11:154-160.
imentally infected cats. J Am Vet Med Assoc. 1997;210:648-650. 58. Lappin MR, Greene CE, Prestwood AK, et al. Diagnosis of recent
36. Brownlee L, Sellon RK. Diagnosis of naturally occurring toxoplas- Toxoplasma gondii infection in cats by use of an enzyme-linked
mosis by bronchoalveolar lavage in a cat. J Am Anim Hosp Assoc. immunosorbent assay for immunoglobulin M. Am J Vet Res.
2001;37:251-255. 1989;50:1580-1585.
37. Alves L, Gorgas D, Vandevelde M, et  al. Segmental meningomy- 59. Lappin MR, Bush DJ, Reduker DW. Feline serum antibody
elitis in 2 cats caused by Toxoplasma gondii. J Vet Intern Med. responses to Toxoplasma gondii and characterization of target
2011;25:148-152. antigens. J Parasitol. 1994;80:73-80.
CHAPTER 72  Toxoplasmosis 703

60. Lappin MR, Powell CC. Comparison of latex agglutination, indi- 72. Lappin M. Unpublished observations, 2012.
rect hemagglutination, and ELISA techniques for the detection of 73. Mitchell SM, Zajac AM, Davis WL, et  al. Efficacy of ponazuril
Toxoplasma gondii-specific antibodies in the serum of cats. J Vet in  vitro and in preventing and treating Toxoplasma gondii infec-
Intern Med. 1991;5:299-301. tions in mice. J Parasitol. 2004;90:639-642.
61. Dabritz HA, Gardner IA, Miller MA, et  al. Evaluation of two 74. Martins-Duarte ES, de Souza W, Vommaro RC. Toxoplasma gon-
Toxoplasma gondii serologic tests used in a serosurvey of domestic dii: the effect of fluconazole combined with sulfadiazine and pyri-
cats in California. J Parasitol. 2007;93:806-816. methamine against acute toxoplasmosis in a murine model. Exp
62. Dubey JP. Duration of immunity to shedding Toxoplasma gondii Parasitol. 2012.
oocysts by cats. J Parasitol. 1995;81:410-415. 75. Dubey JP, Hill DE, Jones JL, et  al. Prevalence of viable Toxo-
63. Cannizzo KP, Lappin MR, Cooper CM, et al. Toxoplasma gondii plasma gondii in beef, chicken, and pork from retail meat stores
antigen recognition by serum IgM, IgG, and IgA of queens and their in the United States: risk assessment to consumers. J Parasitol.
neonatally infected kittens. Am J Vet Res. 1996;57:1327-1330. 2005;91:1082-1093.
64. Burney DP, Lappin MR, Cooper CM, et  al. Detection of Toxo- 76. Gonçalves IN, Uzêda RS, Lacerda GA, et al. Molecular frequency
plasma gondii-specific IgA in the serum of cats. Am J Vet Res. and isolation of cyst-forming coccidia from free ranging chickens in
1995;56:769-773. Bahia State, Brazil. Vet Parasitol. 2012;190:74-79.
65. Lappin MR, George JW, Pedersen NC, et al. Primary and second- 77. Angulo FJ, Glaser CA, Juranek DD, et al. Caring for pets of immu-
ary Toxoplasma gondii infection in normal and feline immunodefi- nocompromised persons. J Am Vet Med Assoc. 1994;205:1711-1722.
ciency virus-infected cats. J Parasitol. 1996;82:733-742. 78. Dabritz HA, Conrad PA. Cats and Toxoplasma: implications for
66. Lappin MR, Greene CE, Dawe DL. Clinical feline toxoplasmosis: public health. Zoonoses Public Health. 2010;57:34-52.
serologic diagnosis and therapeutic management of 15 cases. J Vet 79. Jones JL, Dargelas V, Roberts J, et al. Risk factors for Toxoplasma
Intern Med. 1989;3:139-143. gondii infection in the United States. Clin Infect Dis. 2009;49:878-884.
67. Lappin MR, Dawe DL, Lindl PA, et al. The effect of glucocorticoid 80. Kaplan JE, Benson C, Holmes KK, et al. Guidelines for prevention
administration on oocyst shedding, serology, and cell-mediated and treatment of opportunistic infections in HIV-infected adults
immune responses of cats with recent or chronic toxoplasmosis. and adolescents. Recommendations from CDC, the National
J Am Anim Hosp Assoc. 1992;27:625-632. Institutes of Health, and the HIV Medicine Association of the
68. Colorado State University Veterinary Diagnostic Laboratories. Infectious Diseases Society of America. MMWR Recomm Rep.
http://www.dlab.colostate.edu. Last accessed January 13, 2013. 2009;58(RR04):1-198.
69. Burney DP, Spilker M, McReynolds L, et  al. Detection of Toxo- 81. Wallace MR, Rossetti RJ, Olson PE, et al. Cats and toxoplasmosis
plasma gondii parasitemia in experimentally inoculated cats. risk in HIV-infected adults. JAMA. 1993;269:76-77.
J Parasitol. 1999;5:947-951. 82. Malmasi A, Mosallanejad B, Mohebali M, et  al. Prevention of
70. Lee JY, Lee SE, Lee EG, et al. Nested PCR-based detection of Toxo- shedding and re-shedding of Toxoplasma gondii oocysts in experi-
plasma gondii in German shepherd dogs and stray cats in South Korea. mentally infected cats treated with oral clindamycin: a preliminary
Res Vet Sci. 2008;85:125-127. study. Zoonoses Public Health. 2009;56:102-104.
71. Mitchell SM, Zajac AM, Kennedy T, et al. Prevention of recrudes-
cent toxoplasmic encephalitis using ponazuril in an immunodefi-
cient mouse model. J Eukaryot Microbiol. 2006;53:S164-S165.
CHAPTER 73

Neosporosis
Jane E. Sykes

N. caninum is one of the most important causes of infertility,


Overview of Neosporosis abortion, and neonatal mortality in cattle worldwide. Sheep can
First Described: 1984, in boxer dogs in Norway (Bjerkas and also develop reproductive and neonatal disease, but the eco-
others)1 nomic importance of N. caninum in sheep is less clear when
compared with cattle.4 Herbivores likely become infected when
Cause: Neospora caninum, a coccidial protozoan parasite
they ingest oocysts shed in canine feces or through subclinical
(phylum Apicomplexa)
transplacental transmission. Oocysts sporulate and become
Affected Hosts: Dogs are the definitive hosts and are the only infective 24 to 72 hours after they are shed in canine feces and
species known to complete the sexual phase of N. cani- can survive in the environment for prolonged periods. After
num replication with passage of oocysts in the feces. Clini- oocysts are ingested, sporozoites are released in the intestinal
cal illness primarily occurs in dogs and cattle, but sheep, tract and penetrate intestinal epithelial cells. Organisms can then
deer, and water buffalo may also act as intermediate hosts. disseminate to a variety of tissues, where they ultimately encyst
Geographic Distribution: Worldwide as bradyzoite cysts. In dogs, pregnancy is associated with reac-
tivation of bradyzoite cysts, with replication of tachyzoites and
Mode of Transmission: Dogs can be infected transplacentally subsequent abortion or transplacental transmission. In breeding
or through the transmammary route. Postnatally, dogs dogs, repeated reactivation of N. caninum replication occurs in
may be infected after they ingest infected tissues of inter- late gestation with successive pregnancies, so that multiple litters
mediate hosts or bovine fetal membranes of puppies can be affected. Transmission may also occur via the
Major Clinical Signs: Ascending paralysis, muscle atrophy, transmammary route. Infection of immunologically naïve cattle
neurologic (especially cerebellar) signs, nodular dermati- late in pregnancy is most likely to lead to transplacental spread
tis, respiratory distress with abortion (also known as exogenous transplacental trans-
Differential Diagnoses: Toxoplasmosis, sarcocystosis, hepa- mission).5 However, transplacental transmission as a result of
tozoonosis, deep mycoses, protothecosis, rabies, canine reactivation of latent infection during pregnancy (endogenous
distemper virus infection, West Nile virus infection, canine transplacental transmission) can also occur.6 The pregnant cow
monocytic ehrlichiosis, migrating ascarid infections, neu- typically shows no other overt clinical signs of infection. Although
ronal storage diseases, granulomatous meningoencepha- antibodies to N. caninum have been found in cats and humans,
litis, immune-mediated neuromuscular disease, primary naturally occurring neosporosis has not been described in these
or metastatic neoplasia species.
Aside from transplacental transmission, dogs can become
Human Health Significance: There is no clear evidence that infected and shed oocysts when they ingest bovine placen-
N. caninum infects humans. tal material as well as other bovine tissues, such as muscle,
liver, brain, and heart muscle.7 This horizontal transmission
is important and is thought to maintain infection in the dog
population.8 The DNA of N. caninum has been found in birds,
Etiology and Epidemiology small rodents, and lagomorphs, but whether predation of these
species leads to oocyst shedding by dogs is not clear.4 Experi-
Neospora caninum is an obligately intracellular protozoan mental evidence suggests that ingestion of infected chicken
pathogen that closely resembles Toxoplasma gondii; before its eggs by dogs may lead to oocyst shedding.9 However, inges-
description in 1984, it was misidentified as T. gondii. In contrast tion of oocysts by dogs does not lead to oocyst shedding or
to T. gondii, domestic dogs and wild canids (coyotes, dingos, development of tissue cysts.10 When dogs do shed oocysts,
and probably wolves), and not cats, act as definitive hosts of the shedding begins 5 to 13 days after infection, and only
N. caninum and shed oocysts after ingestion of N. caninum tis- low numbers of oocysts are shed for a short period (several
sue cysts (enteroepithelial cycle) (Figure 73-1). Clinical neospo- days); seroconversion does not always occur. Rarely, pro-
rosis results from systemic replication of N. caninum tachyzoites tracted oocyst shedding (several months) by dogs has been
(which divide by a process known as endodyogeny) and has described.11 Puppies shed more oocysts than adult dogs,12 and
been described in dogs and certain domestic and wild herbivores the number of oocysts shed decreases with repeated exposures
such as cattle, sheep, goats, deer, and marsupials. A related para- to bradyzoite cysts.
site, Neospora hughesi, sporadically causes myeloencephalitis in In dogs, seroprevalence increases with age (because of an
horses.2 This organism has an unknown definitive host that is increased likelihood of exposure through horizontal transmis-
unlikely to be the dog, based on experimental infection studies.3 sion over time) and is higher in free-roaming dogs than in pet

704
CHAPTER 73  Neosporosis 705

FIGURE 73-1  Life cycle of Neospora caninum. Dogs become infected when they ingest tissue cysts in bovine placental material and other bovine tissues. Bradyzoites are released in
the intestine and transform into merozoites, where they undergo merogony. A zygote forms and is shed in the feces as an unsporulated oocyst. In addition, organisms can penetrate the
dog’s intestinal tract and form tissue cysts. Reactivation of these cysts in pregnancy can result in repeated transplacental transmission to the fetus. Herbivores are infected when they ingest
sporulated oocysts. Infection of cattle can lead to transplacental spread of tachyzoites and abortion.

dogs. Dogs that reside on cattle farms or dogs in rural areas Transplacental infection of puppies usually leads to myositis
are also more likely to be seropositive than dogs that reside in and polyradiculoneuritis that initially involves the lumbosacral spi-
urban areas.13-16 Introduction of infected dogs to a farm can nal nerve roots. This is manifested as ascending paralysis, muscle
lead to outbreaks of abortion in cattle on the farm,17 and the atrophy, and fibrous muscle contracture with arthrogryposis (joint
more dogs present on a farm, the greater the chance that cattle contracture), hyperextension of the pelvic limbs, and loss of patel-
will be exposed. Although mixed breed dogs are more likely to lar reflexes.18 Early signs of disease include exercise intolerance,
be seropositive (exposed) than purebred dogs, purebred dogs ataxia, a high-stepping gait, splaying of the pelvic limbs, a “bunny
often develop clinical neosporosis,18 possibly because of genetic hopping” gait, urinary incontinence, and/or muscle pain.18,24-26
defects in cell-mediated immune function. Commonly affected Clinical signs progress in some dogs to lethargy, tetraplegia, inabil-
breeds in the literature and/or in the author’s practice include ity to fully open the mouth, dysphagia, and respiratory difficulty.
boxers, Rhodesian ridgebacks, bull mastiffs, greyhounds, basset Involvement of esophageal muscle may lead to megaesophagus.27,28
hounds, Labrador retrievers, Rottweilers, and West Highland Mandibular paralysis, vomiting, and lethargy were described as the
white terriers. only clinical signs in one affected dog.28 Although ocular lesions
are not a major clinical feature of neosporosis, focal chorioretini-
Clinical Features tis has been reported in some puppies.29 Less often, young dogs
develop signs of hepatitis; pneumonia; meningoencephalomyelitis;
Signs and Their Pathogenesis or myocarditis with arrhythmias, acute onset of respiratory dis-
Although exposure to N. caninum is common, clinical disease tress, or sudden death.26,27,30,31 Puppies with neuromuscular signs
is uncommon in dogs. The development of clinical neosporo- may have subclinical infection of other organ systems.27
sis, the signs that develop, and their rate of progression may In contrast to puppies, adult dogs typically develop polymy-
depend on the virulence of the infecting N. caninum strain and ositis and/or meningoencephalomyelitis, with a variety of neu-
the age and immunocompetence of the host.4,19 Many affected rologic signs.19,27,32 Although lesions can occur throughout the
dogs are infected transplacentally, after which clinical signs central nervous system (CNS),27 N. caninum has a predilection
usually become apparent from 4 weeks to 6 months of age. Up for the cerebellum.19 Some dogs can develop cerebellar atro-
to 50% of puppies in a litter can be infected, but not all puppies phy.19,33 Disseminated infections that involve the myocardium,
develop signs simultaneously, and sometimes only one puppy liver, pancreas, lungs, skin, and/or eyes can also occur in adult
in the litter develops clinical illness.20 Stillbirths and neonatal dogs but are less common than encephalitis. In one unusual
deaths may also be reported.18 Reactivation of subclinical infec- case, N. caninum tachyzoites were found in the peritoneal fluid
tion can occur in older dogs after treatment with immunosup- of a dog with bacterial peritonitis.34
pressive drugs or chemotherapy.21-23 Horizontal acquisition of
infection by adult dogs may occur through consumption of raw Physical Examination Findings
meat; one dog that developed neosporosis had a history of being Physical examination findings in puppies with polyradiculoneuritis-
fed raw elk meat.22 myositis vary from mild ataxia and muscle atrophy to
706 SECTION 4  Protozoal Diseases

TABLE 73-1
Diagnostic Assays Available for Neosporosis in Dogs
Assay Specimen Type Target Performance
Fecal flotation Feces N. caninum oocysts Dogs with clinical neosporosis typically
do not shed oocysts so this has a very
low sensitivity and is not used routinely
for diagnosis. Oocysts cannot be readily
distinguished from those of Hammondia
heydorni.
Fecal PCR assay Feces N. caninum DNA As for fecal flotation.
Cytologic examination CSF, aspirates of skin N. caninum tachyzoites Insensitive except in the case of skin lesions.
lesions, rarely specimens Confirms active infection and is generally
from the respiratory tract associated with disease. Organisms cannot
be readily distinguished from Toxoplasma
gondii.
Serology (IFA or ELISA) Serum or CSF Antibodies to N. caninum Assays can vary in performance between
laboratories. A fourfold increase in titer is
associated with recent infection. Dogs with
chronic neosporosis may not show a rise in
titer, but titers >1:800 in conjunction with
consistent clinical signs suggest neosporosis.
Histopathology Muscle biopsies, multiple N. caninum bradyzoites If organisms are detected in the presence
tissues collected at nec- and tachyzoites of inflammation and necrosis, clinical
ropsy neosporosis is likely. May be difficult to
differentiate N. caninum and T. gondii.
Immunohistochemistry can aid in the
differentiation of the two species.
PCR assay Muscle or skin biopsies, N. caninum DNA Assay performance may vary between
body fluids (especially laboratories. Because positive PCR results
CSF), tissues collected at can occur in healthy dogs, they must be
necropsy interpreted in light of clinical signs and
other findings in the specimens tested.

tetraparesis with rigid extension of one or both pelvic limbs especially cerebellar) signs or nodular dermatitis, especially in
and arthrogryposis. Reduced patellar and sometimes other seg- predisposed breeds or dogs treated with immunosuppressive
mental reflexes (including the perianal reflex) are often pres- drugs. Diagnosis most often relies on serology or detection of
ent.18,24,35,36 Severely affected dogs may be lethargic and exhibit protozoal organisms in body fluids, aspirates, or tissues using
respiratory difficulty, and it may only be possible to open the light microscopy and/or PCR assays (Table 73-1). Often, ante-
mouth a few centimeters. Fever is rare.18 mortem diagnosis is based only on serologic testing because
Adult dogs with N. caninum meningoencephalitis may be organisms are not easily found.
obtunded and exhibit signs that include ataxia, circling, head tilt,
nystagmus, hypermetria, head tremors, delayed placing reactions, Laboratory Abnormalities
abnormal cranial nerve function, anisocoria, depressed segmental Complete Blood Count, Serum Biochemistry Panel, and Urinalysis
reflexes, tetraparesis, cervical hyperesthesia, and seizures. As for toxoplasmosis, there are no specific hematologic or bio-
Dogs with myocardial involvement may develop acute onset chemical abnormalities that suggest neosporosis. The CBC is
of respiratory difficulty and cyanosis.18,27 Cutaneous involvement often normal, but occasionally mild nonregenerative anemia,
is manifested as dermatitis with single or multifocal firm nodules, mild eosinophilia, or monocytosis is evident.18,32 The serum
crusting, and/or ulceration, which can appear at additional sites biochemistry panel reveals increased creatine kinase activity in
over time and may or may not be accompanied by other systemic dogs with myositis.19,23,24,26 Occasionally, increased activity of
signs of infection.21,37-40 Rarely, dogs with pulmonary involvement serum ALT (due to hepatitis or severe myositis) or hyperglobu-
are evaluated for cough or respiratory difficulty.41 linemia is evident.23,32 The urinalysis is generally unremarkable.

Diagnosis Cerebrospinal Fluid Analysis


The CSF of dogs with N. caninum meningoencephalitis may
Neosporosis should be suspected in young dogs (<1 year of be normal or show mild to markedly increased total nuclear
age) that develop ascending paralysis and muscle atrophy. It cell count (TNCC) (sometimes >1000 cells/µL) and mild to
should also be suspected in dogs that develop neurologic (and markedly increased CSF protein concentration.19,32 Usually,
CHAPTER 73  Neosporosis 707

a mononuclear pleocytosis is present, but occasionally the


response is mixed or primarily composed of neutrophils or
eosinophils.19,24,32,35 N. caninum tachyzoites are rarely seen in
CSF.22,32

Diagnostic Imaging
Plain Radiography
Thoracic radiographs in dogs with neosporosis are usually unre-
markable but may show a diffuse interstitial pattern in dogs
with pneumonia, or evidence of megaesophagus in dogs with
neuromuscular signs.18

Sonographic Findings
Abnormalities are rarely detected on abdominal ultrasound
examination of dogs with neosporosis.

Advanced Imaging
MRI of adult dogs with cerebellar signs associated with
N. caninum infection may show moderate to marked bilateral
cerebellar atrophy.19 Meningeal contrast enhancement may be
present.19,32 Single or multifocal T2-weighted and FLAIR hyper-
intensities that enhance with contrast on T1-weighted images
may be present within the cerebellum or other parts of the brain FIGURE 73-2  Sagittal T1-weighted postcontrast MRI image from a 4-year-old
(Figure 73-2). Mild to moderate heterogenous T2-weighted and female spayed Chihuahua with suspected Neospora caninum meningoencephalitis. Mul-
FLAIR hyperintensities can also be seen in the masseter and tiple poorly defined regions of contrast enhancement were identified in the cerebral
temporalis muscles, which may appear atrophied. cortices (large arrow), and mild meningeal enhancement was evident (small arrow). CSF
analysis revealed a mononuclear pleocytosis, and Neospora DNA was detected using PCR
Electrodiagnostic Testing in the cerebrospinal fluid.
Electromyography in dogs with neosporosis may reveal sponta-
neous fibrillation potentials, positive sharp waves, and repetitive
discharges.24,26 In some dogs, nerve conduction studies have
revealed reduced conduction velocities and compound muscle
action potentials.24

Microbiologic Tests
Cytologic Examination
The tachyzoites of N. caninum may be visualized within aspirates
of skin lesions (which often contain many organisms) and rarely
in CSF or other body fluids. They are approximately 6 × 1 µm
and oval to crescent-shaped.22,32,34,37 They may be seen singly,
in pairs, or in larger groups, extracellularly and intracellularly,
and are often associated with a pyogranulomatous inflamma-
tory response (Figure 73-3). In the dog, N. caninum tachyzoites
cannot be distinguished from those of T. gondii on cytologic
examination. Because some cross-reactivity between N. caninum
and T. gondii can occur with immunocytochemistry depending
on the antibodies used, the use of PCR assays is recommended
to definitively identify organisms as N. caninum. For unknown
reasons, such cross-reactivity may be most likely to occur in dermal
neosporosis.8
FIGURE 73-3  Cytology of abdominal fluid sediment in a 7-year old male neutered
Fecal Flotation Rhodesian ridgeback showing intracellular and extracellular Neospora caninum tachyzo-
Fecal flotation with sugar or zinc sulfate solutions can be used ites (arrows). Nucleated cells are predominantly nondegenerate to mildly degenerate
to detect the oocysts of N. caninum in feces. However, clinically neutrophils.
affected dogs do not typically shed oocysts. When oocysts are
seen, they are indistinguishable from those of Hammondia hey- are widely used, but ELISA assays have also been developed and
dorni or Toxoplasma gondii. may be used to detect IgM and IgG antibody. Seroconversion
usually occurs within 2 to 3 weeks of infection, and a rising
Serologic Diagnosis serum titer can be identified in acutely infected dogs.26 This rise
Serologic assays are available commercially that detect antibody may not occur in chronically infected dogs, so in this case a single
to N. caninum in serum and/or in CSF. Titers are often higher high titer in a dog with consistent clinical signs is suggestive
in serum than in the CSF of affected dogs. Indirect IFA assays of the diagnosis. Some dogs with neosporosis have extremely
708 SECTION 4  Protozoal Diseases

A B
FIGURE 73-4  Histopathology of the cerebellum from a 6-year-old intact male Rhodesian ridgeback with neosporosis. A, Large areas of malacia were present admixed with a marked
granulomatous inflammatory infiltrate and abundant protozoal cysts (arrowheads). B, The protozoal cysts had a thick wall and stained positive with anti-Neospora antibodies (not shown).
(Courtesy Dr. Robert Higgins, University of California, Davis Veterinary Anatomic Pathology Service.)

high serum titers (e.g., in the tens of thousands), but others have affected puppies.24 Histopathology in these puppies reveals poly-
only low antibody titers. The latter situation might reflect the radiculoneuritis with myelin loss and axonal degeneration, and
presence of underlying immunodeficiency and inability to pro- there may be evidence of muscle fiber degeneration and fibrosis.
duce antibodies. Because interassay variation may cause results Examination of the brain and spinal cord of dogs with
to differ between laboratories,42 the same laboratory should CNS involvement usually reveals multifocal nonsuppurative
always be used when paired titers are performed or when titers meningoencephalitis and/or myelitis with neuronal degenera-
are monitored over time. tion and necrosis, perivascular cuffing, and gliosis. Both gray
Some weak serologic cross-reactivity may occur between the and white matter can be affected, and the cerebellum may be
antigens of T. gondii and N. caninum, so low titers to T. gon- primarily involved. A mixed inflammatory response with myo-
dii may be present in dogs infected with N. caninum. Cross-­ necrosis and sometimes dystrophic mineralization may be seen
reactivity appears to be less common when IFA assays are in affected skeletal or cardiac muscle. Bradyzoite cysts and/or
used.42 Serologic cross-reactivity can also occur to N. hughesi, tachyzoites may be evident (Figure 73-4). Bradyzoite cysts are
again at a slightly lower titer,43 but the extent to which this up to 100 µm in diameter and have a wall that is 4 µm in diam-
organism infects dogs is not clear. eter, which is thicker than that for T. gondii. Skin biopsies from
dogs with dermal neosporosis contain tachyzoites and pyogran-
Molecular Diagnosis Using the Polymerase Chain Reaction ulomatous inflammation. Occasionally (such as in acute, ful-
Both conventional and real-time PCR assays have been devel- minant infections in immunosuppressed dogs), tachyzoites are
oped for diagnosis of neosporosis.44-46 Some are available for found in other tissues, including the peripheral nerves, the eye,
diagnosis on a commercial basis from veterinary diagnostic reproductive tissues, adrenal glands, lung, liver, kidney, spleen,
laboratories. Suitable specimens for testing include CSF, tissue and esophageal muscle.27 If intralesional protozoa are identi-
biopsies (e.g., muscle), or tissues collected at necropsy. The fied, immunohistochemistry and PCR assays can be used to dis-
sensitivity and specificity of these assays has the potential to tinguish N. caninum from other protozoal species.40 Although
vary considerably between laboratories, and more informa- serologic cross-reactivity between N. caninum and T. gondii is
tion is required on how these assays perform for antemortem uncommon, it can occur with some antibodies, so immunohis-
diagnosis of clinical neosporosis in dogs. Because N. caninum tochemistry is best performed with a panel of antibodies to each
DNA can be detected in tissues from healthy dogs, PCR results protozoan parasite.
must always be interpreted in light of the clinicopathologic In healthy dogs or dogs that die of other diseases, rare brady-
findings and serologic test results. In one study, N. caninum zoite cysts in the absence of an inflammatory response may be
DNA was detected in the liver and/or spleen of one third of found at necropsy in skeletal muscle and the CNS.
shelter dogs that had been euthanized, and PCR assay results
did not always correlate with the results of serology.44 PCR Treatment and Prognosis
assays can be used to confirm that organisms seen in tissues or
body fluids are truly N. caninum and not another protozoan The primary antimicrobial drug that has been used to treat neo-
parasite such as Sarcocystis or T. gondii.23 sporosis in dogs is clindamycin (Table 73-2). Treatment is usually
only temporarily, partially, or completely ineffective, and long
Pathologic Findings periods (>8 weeks) of treatment may be required.18,19,39 Bradyzo-
Gross pathologic findings may be absent in dogs with neospo- ite cysts persist despite treatment with clindamycin.47 Treatment
rosis, or foci of discoloration may be present in the brain and appears to be most effective for cutaneous neosporosis, although
spinal cord, and/or there may be evidence of meningitis (e.g., one dog developed neurologic signs when treatment was discontin-
meningeal thickening or adherence of the meninges to the brain ued, such that reinstitution of long-term treatment was needed.39
or cranial vault). White streaks within muscle may be found in Other treatments that have been successful (or partially successful)
CHAPTER 73  Neosporosis 709

TABLE 73-2
Antimicrobials Used to Treat Neosporosis in Dogs
Drug Dose (mg/kg) Route Interval (hours) Duration (days)
Clindamycin 10-12 mg/kg PO q8h At least 4 weeks
Trimethoprim-sulfa 15 mg/kg PO q12h At least 4 weeks
Pyrimethamine 1 mg/kg PO q24h At least 4 weeks
Ponazuril 20 mg/kg PO q24h At least 4 weeks

are trimethoprim-sulfadiazine-pyrimethamine, combinations of Prevention


clindamycin and trimethoprim-sulfadiazine, or combinations of
clindamycin and pyrimethamine. The efficacy of ponazuril for Neosporosis on farms can be prevented by not allowing dogs
treatment of canine neosporosis requires further study but shows to access bovine placental materials, dead calves, or raw or
promise based on studies in mice and cattle.8 Treatment should be undercooked meat and by preventing dogs from defecating
continued so long as clinical improvement is occurring. The prog- around livestock. Predation should also be discouraged. The
nosis for puppies once muscle contracture has developed is poor. use of a muzzle for dogs that work on farms has been sug-
It has been suggested that once neosporosis has been diagnosed gested to prevent Neospora abortions in cattle.8 It may be
in a puppy, all dogs in a litter should be treated, because the later necessary to stop the breeding of bitches that have puppies
treatment is initiated after the onset of clinical signs, the worse that develop neosporosis, and the owners of puppies that
the prognosis.48 At the minimum, puppies whose littermates have are diagnosed with neosporosis should be advised to inform
succumbed to neosporosis should have their serum antibody titer the breeder of the diagnosis. No drugs are known to prevent
to N. caninum checked, and treatment should be considered for transplacental transmission. Where possible, avoidance of
those with positive titers. The use of immunosuppressive drugs excessive pharmacologic immunosuppression (e.g., with glu-
should be avoided in seropositive puppies. cocorticoids, cyclosporine, azathioprine, or other immuno-
A low dose of glucocorticoids (e.g., prednisone, 0.5 mg/kg PO suppressive drugs) may help to prevent the disease in adult
q12h) in addition to antiprotozoals is often used to treat N. cani- dogs. However, the relative rarity of reactivated neosporosis
num meningoencephalomyelitis, but no prospective controlled in adult dogs does not justify routine serologic screening of
trials have been performed to determine if this alters outcome. Very dogs for infection before commencing immunosuppressive
high doses of methylprednisolone acetate reactivate experimental drug treatment.
N. caninum infections in dogs.49 Physiotherapy may also be
beneficial for young dogs with polyradiculoneuritis-myositis. Public Health Aspects
Immunity and Vaccination Although antibodies to N. caninum have been detected in the
serum of humans, the DNA of N. caninum has never been
Immunity to N. caninum is dependent on cell mediated immune found in humans, and currently N. caninum is not thought to
responses. No vaccines for canine neosporosis exist. be a zoonotic risk.

CASE EXAMPLE U/L), ALT (155 U/L, RR 5-60 U/L), and AST (135 U/L, RR 5-55
U/L). Serum thyroxine concentration was within the reference
range. A urinalysis was unremarkable and aerobic bacterial
Signalment: “Lenny,” a 6-year-old intact male Rhodesian urine culture was negative. Serology for antibodies to Borrelia
ridgeback from coastal northern California burgdorferi, Anaplasma spp., Ehrlichia canis, and antigen of
History: Lenny was evaluated on a referral basis for a 1-week Dirofilaria immitis (4Dx SNAP test, IDEXX Laboratories) was
history of an abnormal gait. The owner first noticed that negative. A T. gondii titer was pending. Lenny was treated
Lenny was not paying attention during an obedience with clindamycin (7.5 mg/kg PO q12h) and doxycycline (5
show. Over the next few days Lenny was observed to mg/kg PO q12h) with no signs of improvement.
stumble on stairs, and he also fell from a bed. For the Lenny had traveled extensively to Iowa, Colorado, and Kentucky
3 days before evaluation the owner had noted that Lenny but had no history of toxin exposure or trauma. He was fed
appeared uncoordinated and off balance. There had been no a raw diet that consisted of turkey necks, chicken meat,
changes in his mentation and no evidence of inappetence, lamb, and vegetables. He was up to date on routine vacci-
vomiting, diarrhea, or changes in thirst or urination. A soft, nations (distemper, adenovirus, parvovirus, and rabies) and
intermittent nonproductive cough had been noted since on heartworm prophylaxis, but did not receive flea or tick
the onset of the other clinical signs. Bloodwork at a local prophylaxis and had been exposed to ticks in the past. He
veterinary clinic showed a normal CBC, increased activity of had been bred once and only one of three puppies survived.
serum creatine kinase (2922 U/L, reference range [RR] 10-200 There was no other significant medical history.
710 SECTION 4  Protozoal Diseases

Physical Examination: ponazuril paste PO q24h) was added. A baseline Schirmer


Body Weight: 40.2 kg. tear test (performed to monitor for keratoconjunctivitis
General: Bright and alert, hydrated. T = 100.5°F (38.1°C), sicca secondary to sulfamethoxazole) was negative. At a
HR = 80 beats/min, panting, mucous membranes pink and recheck examination 3 weeks later, all neurologic signs
moist, CRT 1 s. were persistent but mildly improved. A CBC showed mild,
Musculoskeletal: Body condition score was 4/9. Truncal ataxia nonregenerative anemia (34.9%) and mild bandemia (198
was present with a wide-based stance (see Neurologic cells/µL). Serum CK activity was 88 U/L. CSF analysis showed
Examination). There was no evidence of lameness or muscle a total protein concentration of 33 mg/dL and a TNCC of
atrophy. 2 cells/µL, with 0% eosinophils, 51% small mononuclear
All Other Systems: With the exception of the neurologic cells, and 49% mildly reactive large mononuclear cells. The
examination, no other clinically significant abnormalities serum and CSF antibody titers to N. caninum were both
were detected. 1:5120. A Schirmer tear test was normal. Two weeks later,
Neurologic Examination: The dog was alert and responsive. the trimethoprim-sulfamethoxazole was discontinued.
Truncal ataxia was present with hypermetria that was most Neurologic signs persisted but continued to show mild but
pronounced on the left side. Head tremors were also noted. slow improvement over the next 3 weeks. The ponazuril
A slight left-sided head tilt was present, together with a was discontinued, but after an additional 3 weeks, the head
decreased to absent menace response on the left side. tremors worsened slightly. Blood work was unchanged, CSF
There was normal physiologic nystagmus and no evidence analysis (cisternal and lumbar) was within normal limits, and
of spontaneous nystagmus. Delayed placing reactions serum and CSF N. caninum antibody titers were 1:1280 and
were detected in all four limbs but were most pronounced 1:640, respectively. The prednisone was also discontinued.
on the right side. Spinal reflexes were within normal Two months later (4 months after initial diagnosis), severe
limits. Neuroanatomic location: cerebellar/vestibular ataxia and falling returned over a 2-week period. The owner
disease. declined reinstitution of antiprotozoal treatment and
Laboratory Findings: Muscle biochemistry panel: AST 85 elected euthanasia. A complete necropsy revealed severe,
U/L (21-54 U/L), creatine kinase 1683 U/L (46-320 U/L). locally extensive, necrotizing granulomatous encephalitis
Imaging Findings: that was localized to the cerebellum. Abundant protozoal
Thoracic Radiographs and Abdominal Ultrasound cysts were present within the cerebellar lesions, which were
Examination: No abnormalities were detected. identified using immunohistochemical stains as N. caninum.
Magnetic Resonance Imaging (Brain): On T2-weighted Protozoa were not visualized in any other tissues.
images, there was increased intensity of the vermis and Diagnosis: Granulomatous cerebellitis secondary to
left cerebellar hemisphere. The cerebellum appeared N. caninum infection.
normal on T1-weighted images. Subtle regions of contrast Comments: Neosporosis in this dog resulted from cerebellar
enhancement were suspected in the left cerebellum on encephalitis, which illustrated the suggested predilection
sagittal T1-weighted postcontrast images. of N. caninum for the cerebellum. Development of
Other Testing: CSF analysis: Total nucleated cell count was encephalitis rather than polyradiculoneuritis-myositis is
62 cells/µL (normal <2 cells/µL); total protein was 75 mg/dL typical of neosporosis in adult dogs. It is possible this dog
(normal <25 mg/dL). There were 86% eosinophils, 8% small contracted neosporosis as a result of consumption of a raw
mononuclear cells, 5% large mononuclear cells, and 1% food diet; tissue cysts have been identified in CNS tissues of
nondegenerate neutrophils. The large mononuclear cells were a lamb,50 and chickens are suspected as intermediate hosts
mostly of unremarkable morphology but occasionally had based on seropositivity and positive PCR assay results on
mildly increased cytoplasmic vacuolation. No infectious agents brain tissue.51 The inflammatory response in the CSF was
were seen. profoundly eosinophilic, as previously reported in other
Microbiologic Testing: Serologic testing (IFA): Neospora dogs with neosporosis. Fecal flotation was negative for
caninum serum antibody titer positive at 1:20,480; the CSF N. caninum oocysts, which is expected given the life cycle
titer was 1:640. of the parasite and the short shedding period with low
Zinc sulfate fecal flotation: Negative for parasite ova. numbers of oocysts shed. The fecal examinations were
Fecal Baermann examination: Negative for parasite larvae. performed to evaluate for the possibility of visceral larva
Treatment: Before the Neospora titers were available, a migrans, given the CSF eosinophilia. Although treatment
diagnosis of eosinophilic meningoencephalitis was made, with trimethoprim-sulfamethoxazole and then ponazuril
and the dog was treated with prednisone (0.5 mg/kg led to resolution of CSF abnormalities, clinical signs
PO q12h) but showed no clinical improvement over the showed only mild improvement. Treatment of neosporosis
subsequent 3 days. A second CSF specimen collected at with glucocorticoids remains controversial, but attempts
that time had a cell count of 37 cells/µL and a total protein to reduce the prednisone dose were associated with
concentration of 59 mg/dL, with no change in the differential worsening of clinical signs. Ultimately, all treatment was
cell count. Lenny was discharged from the hospital, and once discontinued because of minimal improvement in clinical
the N. caninum serology results became available (1 week signs but resolution of laboratory abnormalities. Relapse
later), treatment with trimethoprim-sulfamethoxazole (15 subsequently ensued, and the diagnosis was confirmed at
mg/kg PO q12h) and ponazuril (10 mg/kg, or 2.5 mL of 15% necropsy.
CHAPTER 73  Neosporosis 711

SUGGESTED READINGS 19. Garosi L, Dawson A, Couturier J, et al. Necrotizing cerebellitis and
cerebellar atrophy caused by Neospora caninum infection: magnetic
Dubey JP, Lappin MR. Toxoplasmosis and neosporosis. In: Greene CE, resonance imaging and clinicopathologic findings in seven dogs.
ed. Infectious Diseases of the Dog and Cat, 4th ed. St. Louis, MO: J Vet Intern Med. 2010;24:571-578.
Elsevier Saunders; 2012. 20. Heckeroth AR, Tenter AM. Immunoanalysis of three litters born to
Dubey JP, Schares G. Neosporosis in animals—the last five years. Vet a Doberman bitch infected with Neospora caninum. Parasitol Res.
Parasitol. 2011;180:90-108. 2007;100:837-846.
Garosi L, Dawson A, Couturier J, et  al. Necrotizing cerebellitis and 21. Ordeix L, Lloret A, Fondevila D, et al. Cutaneous neosporosis during
cerebellar atrophy caused by Neospora caninum infection: magnetic treatment of pemphigus foliaceus in a dog. J Am Anim Hosp Assoc.
resonance imaging and clinicopathologic findings in seven dogs. J Vet 2002;38:415-419.
Intern Med. 2010;24:571-578. 22. Galgut BI, Janardhan KS, Grondin TM, et al. Detection of Neospora
caninum tachyzoites in cerebrospinal fluid of a dog following pred-
REFERENCES nisone and cyclosporine therapy. Vet Clin Pathol. 2010;39:386-390.
1. Bjerkas I, Mohn SF, Presthus J. Unidentified cyst-forming spo- 23. Fry DR, McSporran KD, Ellis JT, et al. Protozoal hepatitis associ-
rozoon causing encephalomyelitis and myositis in dogs. Z Para- ated with immunosuppressive therapy in a dog. J Vet Intern Med.
sitenkd. 1984;70:271-274. 2009;23:366-368.
2. Pusterla N, Conrad PA, Packham AE, et  al. Endogenous trans- 24. Cuddon P, Lin DS, Bowman DD, et al. Neospora caninum infec-
placental transmission of Neospora hughesi in naturally infected tion in English Springer Spaniel littermates. Diagnostic evaluation
horses. J Parasitol. 2011;97:281-285. and organism isolation. J Vet Intern Med. 1992;6:325-332.
3. Walsh CP, Duncan RB, Zajac AM, et  al. Neospora hughesi: 25. Ishigaki K, Noya M, Kagawa Y, et  al. Detection of Neospora
experimental infections in mice, gerbils, and dogs. Vet Parasitol. caninum–specific DNA from cerebrospinal fluid by polymerase
2000;92:119-128. chain reaction in a dog with confirmed neosporosis. J Vet Med Sci.
4. Dubey JP, Schares G. Neosporosis in animals—the last five years. 2012;74:1051-1055.
Vet Parasitol. 2011;180:90-108. 26. Hay WH, Shell LG, Lindsay DS, et  al. Diagnosis and treatment
5. McCann CM, McAllister MM, Gondim LF, et al. Neospora cani- of Neospora caninum infection in a dog. J Am Vet Med Assoc.
num in cattle: experimental infection with oocysts can result in 1990;197:87-89.
exogenous transplacental infection, but not endogenous trans- 27. Barber JS, Payne-Johnson CE, Trees AJ. Distribution of Neos-
placental infection in the subsequent pregnancy. Int J Parasitol. pora caninum within the central nervous system and other tis-
2007;37:1631-1639. sues of six dogs with clinical neosporosis. J Small Anim Pract.
6. Williams DJ, Hartley CS, Bjorkman C, et al. Endogenous and exog- 1996;37:568-574.
enous transplacental transmission of Neospora caninum—how the 28. Mayhew PD, Bush WW, Glass EN. Trigeminal neuropathy in dogs:
route of transmission impacts on epidemiology and control of disease. a retrospective study of 29 cases (1991-2000). J Am Anim Hosp
Parasitology. 2009;136:1895-1900. Assoc. 2002:262-270.
7. Cavalcante GT, Monteiro RM, Soares RM, et  al. Shedding of 29. Dubey JP, Koestner A, Piper RC. Repeated transplacental trans-
Neospora caninum oocysts by dogs fed different tissues from natu- mission of Neospora caninum in dogs. J Am Vet Med Assoc.
rally infected cattle. Vet Parasitol. 2011;179:220-223. 1990;197:857-860.
8. Reichel MP, Ellis JT, Dubey JP. Neosporosis and hammondiosis in 30. Pumarola M, Anor S, Ramis AJ, et al. Neospora caninum infection in
dogs. J Small Anim Pract. 2007;48:308-312. a Napolitan mastiff dog from Spain. Vet Parasitol. 1996;64:315-317.
9. Furuta PI, Mineo TW, Carrasco AO, et  al. Neospora caninum 31. Meseck EK, Njaa BL, Haley NJ, et  al. Use of a multiplex poly-
infection in birds: experimental infections in chicken and embryo- merase chain reaction to rapidly differentiate Neospora caninum
nated eggs. Parasitology. 2007;134:1931-1939. from Toxoplasma gondii in an adult dog with necrotizing myocar-
10. Bandini LA, Neto AF, Pena HF, et  al. Experimental infection of ditis and myocardial infarct. J Vet Diagn Invest. 2005;17:565-568.
dogs (Canis familiaris) with sporulated oocysts of Neospora cani- 32. Gaitero L, Anor S, Montoliu P, et al. Detection of Neospora cani-
num. Vet Parasitol. 2011;176:151-156. num tachyzoites in canine cerebrospinal fluid. J Vet Intern Med.
11. McGarry JW, Stockton CM, Williams DJ, et al. Protracted shedding 2006;20:410-414.
of oocysts of Neospora caninum by a naturally infected foxhound. 33. Lorenzo V, Pumarola M, Siso S. Neosporosis with cerebellar
J Parasitol. 2003;89:628-630. involvement in an adult dog. J Small Anim Pract. 2002;43:76-79.
12. Gondim LF, McAllister MM, Gao L. Effects of host maturity and 34. Holmberg TA, Vernau W, Melli AC, et  al. Neospora caninum
prior exposure history on the production of Neospora caninum associated with septic peritonitis in an adult dog. Vet Clin Pathol.
oocysts by dogs. Vet Parasitol. 2005;134:33-39. 2006;35:235-238.
13. Antony A, Williamson NB. Prevalence of antibodies to Neospora 35. Peters M, Wagner F, Schares G. Canine neosporosis: clinical and
caninum in dogs of rural or urban origin in central New Zealand. pathological findings and first isolation of Neospora caninum in
N Z Vet J. 2003;51:232-237. Germany. Parasitol Res. 2000;86:1-7.
14. Basso W, Venturini L, Venturini MC, et al. Prevalence of Neospora 36. Ruehlmann D, Podell M, Oglesbee M, et  al. Canine neosporo-
caninum infection in dogs from beef-cattle farms, dairy farms, and sis: a case report and literature review. J Am Anim Hosp Assoc.
from urban areas of Argentina. J Parasitol. 2001;87:906-907. 1995;31:174-183.
15. Sicupira PM, de Magalhaes VC, Galvao Gda S, et al. Factors asso- 37. Boyd SP, Barr PA, Brooks HW, et al. Neosporosis in a young dog
ciated with infection by Neospora caninum in dogs in Brazil. Vet presenting with dermatitis and neuromuscular signs. J Small Anim
Parasitol. 2012;185:305-308. Pract. 2005;46:85-88.
16. Bruhn FR, Figueiredo VC, Andrade Gda S, et  al. Occurrence of 38. Poli A, Mancianti F, Carli MA, et al. Neospora caninum infection
anti–Neospora caninum antibodies in dogs in rural areas in Minas in a Bernese cattle dog from Italy. Vet Parasitol. 1998;78:79-85.
Gerais, Brazil. Rev Bras Parasitol Vet. 2012;21:161-164. 39. McInnes LM, Irwin P, Palmer DG, et al. In vitro isolation and char-
17. Dijkstra T, Barkema HW, Hesselink JW, et al. Point source exposure acterisation of the first canine Neospora caninum isolate in Australia.
of cattle to Neospora caninum consistent with periods of common Vet Parasitol. 2006;137:355-363.
housing and feeding and related to the introduction of a dog. Vet 40. Perl S, Harrus S, Satuchne C, et al. Cutaneous neosporosis in a dog
Parasitol. 2002;105:89-98. in Israel. Vet Parasitol. 1998;79:257-261.
18. Barber JS, Trees AJ. Clinical aspects of 27 cases of neosporosis in 41. Greig B, Rossow KD, Collins JE, et al. Neospora caninum pneumo-
dogs. Vet Rec. 1996;139:439-443. nia in an adult dog. J Am Vet Med Assoc. 1995;206:1000-1001.
712 SECTION 4  Protozoal Diseases

42. Silva DA, Lobato J, Mineo TW, et al. Evaluation of serological tests 47. Dubey JP, Vianna MC, Kwok OC, et al. Neosporosis in Beagle dogs:
for the diagnosis of Neospora caninum infection in dogs: optimiza- clinical signs, diagnosis, treatment, isolation and genetic character-
tion of cut off titers and inhibition studies of cross-reactivity with ization of Neospora caninum. Vet Parasitol. 2007;149:158-166.
Toxoplasma gondii. Vet Parasitol. 2007;143:234-244. 48. Dubey JP, Lappin MR. Toxoplasmosis and neosporosis. In: Greene
43. Gondim LF, Lindsay DS, McAllister MM. Canine and bovine CE, ed. Infectious Diseases of the Dog and Cat, 4th ed. St. Louis,
Neospora caninum control sera examined for cross-reactivity using MO: Elsevier Saunders; 2012.
Neospora caninum and Neospora hughesi indirect fluorescent anti- 49. Dubey JP, Lindsay DS. Neosporosis in dogs. Vet Parasitol.
body tests. J Parasitol. 2009;95:86-88. 1990;36:147-151.
44. Ghalmi F, China B, Kaidi R, et  al. Detection of Neospora cani- 50. Dubey JP, Hartley WJ, Lindsay DS, et  al. Fatal congenital Neos-
num in dog organs using real time PCR systems. Vet Parasitol. pora caninum infection in a lamb. J Parasitol. 1990;76:127-130.
2008;155:161-167. 51. Costa KS, Santos SL, Uzeda RS, et al. Chickens (Gallus domesticus)
45. Langoni H, Matteucci G, Medici B, et al. Detection and molecular are natural intermediate hosts of Neospora caninum. Int J Parasi-
analysis of Toxoplasma gondii and Neospora caninum from dogs with tol. 2008;38:157-159.
neurological disorders. Rev Soc Bras Med Trop. 2012;45:365-368.
46. Schatzberg SJ, Haley NJ, Barr SC, et al. Use of a multiplex polymerase
chain reaction assay in the antemortem diagnosis of toxoplasmosis
and neosporosis in the central nervous system of cats and dogs. Am J
Vet Res. 2003;64:1507-1513.
CHAPTER 74

Leishmaniosis
Jane E. Sykes, Gad Baneth, and Christine A. Petersen

CANINE LEISHMANIOSIS
Overview of Leishmaniosis
First Described: The cause of visceral leishmaniosis was first Etiology and Epidemiology
described in India in 1903 (separately by William Leishman
and Charles Donovan)1 The leishmaniases are a group of zoonotic vector-borne diseases
Causes: Canine and feline visceral leishmaniosis is caused by caused by various species of the protozoa Leishmania, which are
Leishmania infantum (syn. L. chagasi) (phylum Euglenozoa, primarily transmitted by sandflies. Around 30 species of Leish-
class Kinetoplasta, family Trypanosomatidae). Other spe- mania exist worldwide, and at least 20 species cause disease in
cies such as L. braziliensis cause localized cutaneous lesions humans, although the taxonomy of Leishmania spp. has been
in South America (American tegumentary leishmaniosis). highly debated.2,3 There are two subgenera of Leishmania, Leish-
mania and Vianna, which develop in the foregut or the hindgut/
Affected Hosts: Dogs and less commonly cats; also humans,
midgut of the sandfly vector, respectively. Human and animal dis-
rodents, wild canids, and marsupial species
ease due to Leishmania spp. is distributed across five continents
Geographic Distribution: Primarily southern Europe and the (Africa, Asia, Europe, and the Americas) and nearly 100 countries
Middle East, central and South America; also some parts (Figure 74-1). Disease syndromes in humans are divided into cuta-
of the United States. Travel-related disease can occur in neous leishmaniasis (CL), mucosal leishmaniasis (ML), and visceral
non-endemic regions worldwide. leishmaniasis (VL). Disease syndromes in dogs are divided into
Mode of Transmission: Phlebotomus and Lutzomyia sandflies. visceral leishmaniosis and American tegumentary leishmaniosis, a
Transmission through blood transfusion as well as vertical localized cutaneous form that occurs in parts of South America.
transmission can occur. The term ‘leishmaniosis’ as opposed to ‘leishmaniasis’ is preferred
to distinguish the disease in animals from that in humans.
Major Clinical Signs: Weight loss, inappetence, scaling
Leishmania species are transmitted by female sandflies of
and/or ulcerative cutaneous lesions, onychogryphosis,
the genus Phlebotomus in the Old World (Africa, Asia, and
fever, keratoconjunctivitis, uveitis, lymphadenopathy,
Europe) and Lutzomyia in the New World (i.e., the Americas).
hepatosplenomegaly, pallor, lameness, signs of renal
The regional prevalence of leishmaniasis, and the type of disease
failure
syndrome that develops, varies with the infecting Leishmania
Differential Diagnoses: Differential diagnoses in dogs with species present, sandfly species in the region, and host responses
visceral disease include canine monocytic ehrlichio- to infection. Some sandfly species transmit certain Leishmania
sis, babesiosis, histoplasmosis, brucellosis, hemic species primarily to humans (anthroponotic leishmaniasis),
neoplasia, and primary autoimmune diseases such as whereas other sandfly species transmit Leishmania species to
systemic lupus erythematosus. Differential diagnoses mammalian host species, such as dogs, cats, and rodents. Dogs
for cutaneous lesions in dogs include demodectic or and rodents are important reservoirs for Leishmania infection of
sarcoptic mange, pyoderma, Malassezia dermatitis, humans (zoonotic leishmaniasis). Thus, the major mammalian
cutaneous vasculitis, and pemphigus foliaceus. Differ- reservoirs for the disease in a region depend on the Leishmania
ential diagnoses for cutaneous lesions in cats include species and the sandfly species present.
squamous cell carcinoma, cutaneous vasculitis, her- The life cycle of Leishmania alternates between two major
pesviral dermatitis, and dermatophytosis. The primary forms: the promastigote and the amastigote (Figure 74-2). The
differential for American tegumentary leishmaniasis is promastigote resides in the gut of the sandfly vector and is an
sporotrichosis. elongated, flagellated form. Promastigotes are inoculated into
Human Health Significance: Human infections are primarily tissues when the sandfly feeds, where they are phagocytized
transmitted by sandflies and involve a large number of dif- by macrophages in the dermis and transform into intracellu-
ferent Leishmania species. Dogs are considered the major lar, nonmotile, ovoid, or round amastigotes. The amastigotes
reservoir for L. infantum infections. Precautions should survive and replicate within macrophage phagolysosomes, and
be taken when handling potentially infected tissue and eventually the infected macrophages rupture. Released amasti-
blood specimens. gotes then infect other macrophages that have been attracted
to the site of infection. If the infected host fails to control the
infection, amastigotes disseminate via regional lymphatics and
the blood to infect the entire reticuloendothelial system. Intra-
cellular amastigotes that are ingested by a female sandfly during

713
714 SECTION 4  Protozoal Diseases

FIGURE 74-1  Geographic distribution of visceral leishmaniases in humans and dogs.

feeding convert back into promastigotes over a period of about


1 week, and replication of promastigotes within the sandfly
completes the life cycle. Sandflies breed in cracks in the walls
of dwellings, rubble, and in rodent burrows, feed primarily at
night, and fly a maximum of 2.5 km from their breeding sites.
As a result, the prevalence of infection in dogs can vary consid-
erably between adjacent regions. Lesser modes of transmission
of Leishmania spp. include vertical transmission from bitches to
puppies, venereal transmission, and blood transfusion.4-7 These
modes of transmission become more obvious in regions where
suitable sandfly vectors do not exist. Aggressive interactions
between dogs also have the potential to transmit infection, but
this route has not been proven.
Canine visceral leishmaniosis in dogs and cats is caused by
Leishmania infantum (also referred to as Leishmania chagasi
in Central and South America). L. infantum is endemic in the
Mediterranean basin, the Middle East, and Asia and may have
been introduced to Latin America by early explorers and their
domestic dogs.8 Because leishmaniosis has a long incubation
period and prolonged disease course, travel-related leishmaniosis
in dogs and cats is diagnosed sporadically in regions where sand-
fly transmission does not occur, such as parts of Scandinavia,
Switzerland, Japan, the United Kingdom, and the western United
States (most often in dogs imported from southern Europe).9-13
In highly endemic regions of the Mediterranean and Brazil,
FIGURE 74-2  Life cycle of Leishmania infantum. Dogs, cats, and humans become infection rates among dogs vary widely but may reach 80%.14-17
infected when they are bitten by a sandfly. Promastigotes are inoculated into the der- At least 2.5 million dogs are infected in southwestern Europe
mis and penetrate macrophages. Within the macrophage, they transform into amas- alone.14 The disease may be spreading northward in Europe, and
tigotes and replicate within a phagolysosome. The macrophage ruptures and new has recently appeared in nontraveled dogs in countries such as
macrophages are infected. If the host fails to control the infection in the skin, amasti- Germany and Hungary.5,18,19 Spread of Leishmania spp. into
gotes disseminate via regional lymphatics and the blood to infect the entire reticulo- new areas can occur when humans bring infected dogs into
endothelial system. Amastigotes that are ingested by a female sandfly during feeding regions where competent insect vectors exist, or when sandfly
convert back into promastigotes, and replication of promastigotes within the sandfly populations expand as a result of factors such as climate change.
completes the life cycle.
Risk factors for infection in dogs include age at least 2 years,
prolonged exposure to the outdoors, lack of topical insecticide
use, and, in some studies, a short haircoat.20-23 Some authors have
described a bimodal age distribution that peaks at 3 years and at
CHAPTER 74  Leishmaniosis 715

greater than 8 years of age.24 Sex is not an important risk factor


for infection. Factors that promote proliferation of sandfly popula-
tions, such as poor sanitation and limited garbage collection, as
well as the occurrence of stray dogs around homes, also put dogs at
risk of infection.25 In Europe, German shepherd dogs, boxers, Rott-
weilers, cocker spaniels, and Dobermans may be at increased risk
for leishmaniosis; the Ibizan hound in Spain appears to be resistant
to clinical disease.24,26-29 Concurrent diseases are common in dogs
with leishmaniosis in endemic regions and include transmissible
venereal tumor,30 demodicosis,31 sarcoptic mange,32 and a variety
of other vector-borne diseases such as canine monocytic ehrlichio-
sis, babesiosis, hepatozoonosis, and dirofilariasis.32-36 Concurrent
lymphoma has also been described.32
Uncommonly, dogs worldwide are infected by other spe-
cies of Leishmania, which include Leishmania donovani,
Leishmania tropica, Leishmania braziliensis, Leishmania
peruviana, Leishmania mexicana, Leishmania panamensis,
Leishmania guyanensis, Leishmania amazonensis, and other
as-yet-uncharacterized species.37-45 L. braziliensis, L. guyanensis,
and L. panamensis have been isolated from dogs with Ameri-
can tegumentary leishmaniosis. Outbreaks of American tegu-
mentary leishmaniosis have occurred in humans and dogs.43

Canine Leishmaniosis in North America


Most sporadic cases of visceral leishmaniosis in dogs in North
America are diagnosed in dogs with a travel history to other con-
tinents. Infected dogs continue to be introduced every year as a FIGURE 74-3  Periocular alopecia and crusting in an 8-year-old male neutered husky
result of travel from endemic regions in Europe and Latin America. mix with visceral leishmaniosis. The dog was evaluated in northern California but had a
In addition, over the past 30 years, visceral disease has been travel history to Spain. Lesions are also present on the muzzle and the pinnae.
described in dogs in the United States that have had no apparent
travel history. These have included a basenji dog from a kennel
in Texas and dogs of foxhound breed from Ohio, Oklahoma, Cutaneous lesions most commonly manifest as alopecia,
and Colorado.46-49 Involvement of multiple dogs in a foxhound scaling, and/or ulceration but can be nodular or papular (Figure
kennel was reported in Oklahoma,47 and outbreaks in foxhounds 74-3). They are often noticed before signs of systemic infection
have also been described in New York, Alabama, and Michigan.50 develop, despite the fact that they occur as a consequence of
Spread to other dogs has occurred as a result of transplacental parasite dissemination. Many dogs also develop onychogryphosis
transmission and, in rare cases, transfusion of infected blood.4,6 (abnormally long or brittle claws).
A competent arthropod vector for Leishmania spp. has not yet Systemic signs of visceral leishmaniosis include fever, weight
been identified in the United States, and infection has not been loss, muscle atrophy, inappetence, and lethargy; oral ulceration;
detected in surveys of non-foxhound dogs in the United States.51 progressive splenomegaly and lymphadenomegaly; mucosal
pallor due to anemia; and rarely, hepatomegaly (Table 74-1).
Clinical Features Development of autoantibodies and circulating immune com-
plexes is thought to lead to immune-mediated thrombocytope-
Signs and Their Pathogenesis nia and/or thrombocytopathia and signs such as epistaxis or
The outcome of Leishmania spp. infection is variable. Some dogs melena, lameness and joint swelling due to immune-mediated
completely eliminate the infection, and a small percentage of dogs polyarthritis, myositis, uveitis, vasculitis, and glomerulone-
develop severe, life-threatening disease. Some dogs remain persis- phritis.57-62 Positive antinuclear antibody tests are found in up
tently but subclinically infected, with the possibility of reactivation to 50% of dogs tested.32,61,63 Many dogs also have positive
later in life with immunosuppression; these dogs also act as reser- Coombs’ tests.32,63
voir hosts and can infect sandflies.52 In general, dogs that mount Glomerular injury ultimately culminates in nephrotic syn-
strong T-cell–mediated immune responses eliminate the infection, drome and/or failure of tubular function for many dogs, with
whereas those that mount weak cell-mediated immune responses polyuria and polydipsia, vomiting, diarrhea, and dehydration.
and marked, nonprotective humoral (IgG) immune responses Renal failure may occur in the absence of other lesions.32 Anemia
develop clinical disease.53 Host genetic factors are important in may be due to a combination of factors that include blood loss,
determining progression to clinical disease, and several candidate inflammation, renal failure, immune-mediated destruction, and
genes have been identified in dogs that may influence susceptibil- marrow aplasia or hypoplasia. In addition to uveitis, other ocu-
ity.28,54-56 When clinical disease appears, it occurs after a long lar lesions include blepharitis, conjunctivitis, panophthalmitis,
incubation period (up to 7 years) and varies from mild papular or and keratoconjunctivitis. Keratoconjunctivitis results from direct
exfoliative dermatitis to severe disseminated disease due to mas- infection of the lacrimal gland by Leishmania.64
sive proliferation of histiocytes, B lymphocytes, and plasma cells Atypical presentations of canine visceral leishmaniosis have
in reticuloendothelial tissues and immune-mediated consequences also been reported. Almost any organ can be affected, with
of chronic, persistent infection. reports of Leishmania prostatitis and infertility65; nodular lesions
716 SECTION 4  Protozoal Diseases

TABLE 74-1
Clinical Signs in Dogs with Visceral Leishmaniosis
Clinical Sign Prevalence (%)
Lymphadenomegaly 65-90
Cutaneous lesions 51-89
Weight loss 25-64
Lethargy 11-60
Pallor 58
Splenomegaly 10-53
Polydipsia 4-40
Abnormal locomotion or polyarthritis 3-38
Pyrexia 4-36
Onychodystrophy 20-31
Diarrhea 30
Vomiting 26
Ocular lesions (keratoconjunctivitis, 7-24
uveitis, panophthalmitis, blepharitis)
Inappetence 17 FIGURE 74-4  Poor body condition of 8-year-old male intact foxhound that was
Polyphagia 15 seropositive and PCR positive for Leishmania infantum. (Courtesy Dr. Christine Petersen,
Iowa State University.)
Epistaxis 6-15
Melena 13
Onychogryphosis may be present. Lameness or reluctance to
Data from Ciaramella P, Oliva G, Luna RD, et al. A retrospective clini- move may be associated with joint swelling, crepitus, and pain.72
cal study of canine leishmaniasis in 150 dogs naturally infected by Leish- Epistaxis may also be noted.
mania infantum. Vet Rec 1997;141:539-543; Slappendel RJ. Canine
leishmaniasis. A review based on 95 cases in The Netherlands. Vet Q
1988;10:1-16; Koutinas AF, Carlotti DN, Koutinas C, et al. Claw his- Diagnosis
topathology and parasitic load in natural cases of canine leishmaniosis
associated with Leishmania infantum. Vet Dermatol 2010;21:572-577. Diagnosis of leishmaniosis may be straightforward, such as
when organisms are seen cytologically in impression smears of
typical skin lesions, or it may require assessment of the results of
of the tongue and other mucosal surfaces66,67; pancreatitis68; gas- a combination of diagnostic assays in light of clinicopathologic
trointestinal tract, myocardial, or pulmonary involvement69-71; findings (Table 74-2). Specific diagnostic assays include cytologic
osteomyelitis72,73; meningitis74; and granuloma formation within or histopathologic examination of affected tissues, serology, cul-
the spinal cord.75 ture of the organism, or the results of PCR assays on affected
tissues or blood. In endemic areas, diagnosis may be challenging
American Tegumentary Leishmaniosis (1) because dogs may be infected with the parasite for long peri-
American tegumentary leishmaniosis in dogs is characterized by ods of time in the absence of clinical signs, so the significance of
development of a nodular skin lesion at the site of a sandfly bite positive test results may be unclear, and (2) because co-infections
that enlarges and eventually may ulcerate. Lesions most often with other vector-borne pathogens may complicate the clinical
occur on regions of the body that have less hair, such as the picture and cause signs that resemble those of leishmaniosis.
nose, perineal region, scrotum, and pinnae.43,76
Laboratory Abnormalities
Physical Examination Findings Complete Blood Count
Physical examination findings in dogs with visceral disease The most common CBC abnormality in dogs with leishmaniosis
include lethargy, muscle atrophy or cachexia, variable and is a mild to moderate, normocytic, normochromic, nonregen-
typically low-grade fever, focal or generalized peripheral erative anemia. Mild thrombocytopenia is present in up to 50%
lymphadenomegaly, and hepatosplenomegaly on abdominal of affected dogs.32,63,77 Total leukocyte, neutrophil, lympho-
palpation (Figure 74-4). Lymphadenomegaly can be severe and cyte, monocyte, and eosinophil counts may be decreased, within
be suggestive of lymphoma. Although not always detected, skin the reference range, or increased. Pancytopenia may be present.
lesions consist of scaling and crusting, hyperkeratosis, alope- Although most dogs are lymphopenic, some have moderate to
cia, erythema, and cutaneous ulceration and may be nonpru- severe lymphocytosis.77
ritic or pruritic. They most often occur on the limbs, pinnae,
muzzle, and periocular regions. Mucocutaneous lesions can Serum Biochemical Tests
also occur. Ocular involvement may be associated with muco- Changes on serum biochemistry analysis in dogs with leish-
purulent ocular discharge, blepharitis, and anterior uveitis. maniosis include hyperglobulinemia, hypoalbuminemia, and
CHAPTER 74  Leishmaniosis 717

TABLE 74-2
Diagnostic Assays Currently Available for Leishmaniosis in Dogs and Cats
Assay Specimen Type Target Performance
Cytologic Aspirates of lymph nodes, Leishmania infantum In the absence of experience, organisms
examination spleen, or bone marrow; amastigotes within may be confused with fungi such as
impression smears of skin mononuclear phagocytes Histoplasma capsulatum or Sporothrix
­lesions; rarely blood smears schenckii. Organisms may be few in
or buffy-coat preparations number and in some cases are absent.
Leishmania culture Aspirates or biopsies of L. infantum Sensitive and specific but requires special
affected tissues techniques and expertise that are not
widely available.
Antibody serology Serum Antibodies to L. infantum Most infected dogs test positive (sensi-
(IFA or ELISA) tivity > 90%), but positive test results
occur in healthy animals, so results
must be interpreted in light of clinical
signs. Sensitivity may be lower in cats.
False positives can occur in dogs
exposed to Trypanosoma spp. (IFA)
and dogs vaccinated with Leishmania
vaccines.
Real-time PCR Conjunctival swabs, aspirates L. infantum DNA May be useful in dogs with suggestive
assays of affected tissues, lymph lesions in which organisms cannot be
node, bone marrow, whole found cytologically. Sensitivity and
blood, skin biopsies specificity depends on assay design.
Quantitative PCR may be useful for
evaluation of treatment efficacy.
Histopathology Biopsy specimens from L. infantum amastigotes Use of immunohistochemistry may
affected tissues (e.g., skin increase sensitivity and can be used to
lesions) confirm the presence of Leishmania spp.
in tissues.

IFA, Immunofluorescent antibody.

mild to moderate azotemia.32,63,77 Hyperglobulinemia and and increased numbers of lymphocytes and plasma cells.79,80
hypoalbuminemia are present in more than 75% of affected Amastigotes may also be visible.
dogs. Serum globulin concentration ranges from mild to
severe (up to 8 g/dL) and reflects a polyclonal gammopa- Diagnostic Imaging
thy; monoclonal gammopathy is rare.78 Azotemia has been Abdominal radiography or ultrasonography in dogs or cats
reported in 16% to 38% of dogs. Some studies have shown with visceral leishmaniosis can reveal abdominal lymphadeno-
mild to moderate increases in liver enzyme activities. megaly, splenomegaly, and hepatomegaly. Radiographic lesions
of long bones in dogs with bone involvement consist of soft
Urinalysis periosteal proliferation, increased or decreased intramedullary
Dogs with leishmaniosis may have proteinuria or isosthenuria, opacity, and/or cortical and medullary destruction.63,72 Lesions
secondary to glomerular and tubulointerstitial renal function tend to be bilateral and symmetrical. Joint radiographs in dogs
impairment, respectively. Cylindruria may be detected. with signs of arthritis may reveal erosive changes, or arthritis
may be non-erosive.
Synovial Fluid Examination
Cytologic examination of synovial fluid from dogs with Microbiologic Tests
Leishmania polyarthritis reveals increased numbers of neu- Cytologic Examination
trophils and/or lymphocytes.72 In some cases, Leishmania Identification of Leishmania spp. amastigotes in lesions with
amastigotes are visible within macrophages in the synovial cytology is diagnostic for leishmaniosis (Figure 74-5). Organ-
fluid. ism densities are particularly high in skin lesions, bone marrow,
and the spleen, but specimens collected from other tissues, such
Bone Marrow Examination as lymph node and hepatic aspirates, may also contain organ-
Bone marrow cytologic alterations described in canine leish- isms. Organisms are not found in some lesions, and inexperi-
maniosis include erythrophagocytosis, erythroid hypoplasia and enced microscopists may fail to identify organisms when present
dysplasia, eosinophilic hypoplasia, granulocytic hyperplasia, or confuse them with other parasites, such as Trypanosoma,
718 SECTION 4  Protozoal Diseases

Histoplasma, or Sporothrix schenckii. Rarely, organisms are Molecular Diagnosis Using the Polymerase Chain Reaction
seen in circulating white blood cells.81 Several different conventional and real-time PCR assays have
been developed and evaluated for detection of Leishmania spp.
Serologic Diagnosis in dogs and cats. Real-time PCR assays that detect Leishma-
Serologic assays based on immunofluorescent antibody (IFA), nia spp. are now offered on a commercial basis by some veteri-
ELISA, Western immunoblot, and agglutination techniques nary diagnostic laboratories worldwide. Suitable specimens for
have all been used for serodiagnosis of canine leishmaniosis, testing include splenic, hepatic, lymph node, or bone marrow
or for screening apparently healthy dogs in endemic areas for aspirates; whole blood; skin biopsies; conjunctival swabs; and
evidence of infection. IFA is commonly used by veterinary even oral swabs. Conjunctival swabs; aspirates of spleen, bone
practitioners in Europe82 and has a diagnostic sensitivity that marrow, or lymph nodes; and skin biopsies are most likely to
ranges from 90% to 100% and a specificity of 80% to 100% yield positive results.85,86 Positive PCR assay results in endemic
in dogs that have not received a Leishmania vaccine. Dogs areas do not confirm that disease is caused by Leishmania spp.,
with Trypanosoma spp. infections may have false-positive test because dogs without clinical signs can test positive.87 However,
results due to serologic cross-reactivity between Trypanosoma in one study, clinical disease subsequently appeared in 71% of
and Leishmania antigens. ELISA assays based on recombinant ELISA-positive dogs that lacked clinical signs but had medium
antigens often have higher specificity and are technically less to high parasite loads (more than 10 parasites/mL of blood) as
difficult to perform than IFA.42,83 A rapid lateral flow assay is determined with a quantitative PCR assay.86 PCR assays may
available commercially (SNAP Canine Leishmania Antibody also be useful in order to monitor the efficacy of treatment.
Test Kit, IDEXX Laboratories) that had an overall sensitivity
of 95% and a specificity of 91% in one study of 400 dogs from Culture
Brazil.83 Other immunochromatographic assays are available Leishmania spp. can be isolated in culture from specimens
that vary in their sensitivity. such as lymph nodes, skin biopsies, spleen, or bone marrow,
Negative serologic test results may reflect delayed seroconver- although special media and expertise are required, and incuba-
sion in some animals, which can take several months to occur. tion times of up to 30 days may be needed.
However, many dogs with clinical signs have already serocon-
verted because of the long incubation period. Importantly, in Pathologic Findings
endemic areas, positive serologic test results do not imply that Gross Pathologic Findings
disease is due to Leishmania spp. infection, because subclini- Gross pathologic findings in dogs with visceral leishmaniosis
cal infections are widespread. Thus, for diagnosis of clinical include cachexia; generalized enlargement of the spleen, lymph
leishmaniosis, results must be interpreted in conjunction with nodes, and sometimes the liver; focal pale nodular lesions in a
clinical findings and cytologic and histopathologic evaluation variety of organs; bone and joint lesions; and gastrointestinal
of affected tissues. Because seroconversion occurs in dogs that ulcerations.
have received the Leishmania vaccine, current serologic assays
cannot be used to efficiently differentiate between infected and Histopathologic Findings
vaccinated animals.84 Histopathology of most affected tissues in dogs with leish-
maniosis typically reveals granulomatous to pyogranuloma-
tous and lymphoplasmacytic inflammation, with variable
numbers of intrahistiocytic amastigotes (Figure 74-6). Skin
lesions are accompanied by orthokeratotic to parakeratotic

FIGURE 74-5  Bone marrow aspirate cytology findings in a dog with visceral leish- FIGURE 74-6  Histopathology of the spleen in a dog with visceral leishmaniosis. Mul-
maniosis. Numerous Leishmania amastigotes can be seen within a histiocyte. (Courtesy tiple amastigotes (which have a granular appearance, arrows) are present within macro-
Dr. Sheri Ross, University of California, Davis – San Diego.) phages. H&E stain; bar = 20 µm. (Courtesy Dr. Christine Petersen, Iowa State University.)
CHAPTER 74  Leishmaniosis 719

hyperkeratosis, acanthosis, and ulceration. Claw histopathol- sodium stibogluconate; allopurinol; amphotericin B; and miltefos-
ogy in dogs with onychogryphosis reveals lichenoid mono- ine (Table 74-3). Ketoconazole has also been used. Unfortunately,
nuclear cell infiltration, with or without basal keratinocyte regardless of the treatment used, complete parasitologic cure is
vacuolation and dermo-epidermal clefting; organisms are rare, and relapses are frequent.93,94 In endemic areas, reinfec-
generally not visible.88 Renal lesions consist of membranop- tions occur and contribute to apparent treatment failures. Staging
roliferative glomerulonephritis, lymphoplasmacytic tubuloint- systems and consensus treatment guidelines have been published
erstitial nephritis, and rarely amyloidosis (Figure 74-7).46,89-91 (Table 74-4).29,95
Immunohistochemistry can be used to unambiguously detect Pentavalent antimonials are thought to inhibit protozoal
Leishmania amastigotes in tissue sections. Immunohistochem- enzymes and damage protozoal DNA, whereas allopuri-
istry may permit detection of Leishmania spp. when organisms nol interferes with protein synthesis by Leishmania spp. (see
are not seen with routine histopathology.70,92 In situ hybrid- Chapter 10 for more information on these drugs). In general,
ization has also been used to identify Leishmania amastigotes a combination of meglumine antimoniate (100 mg/kg SC
in paraffin-embedded specimens.92 q24h for 4 weeks) and allopurinol (10 mg/kg PO q12h until
clinical signs have resolved and quantitative serology becomes
Treatment and Prognosis negative) is the treatment of choice for dogs with leishmani-
osis.96 Clinical improvement generally occurs within 1 to 2
Antiprotozoal Therapy months of treatment initiation if no overt renal disease is pres-
Drugs with activity against Leishmania spp. include the pen- ent. Complete or near-complete clinical remission rates of
tavalent antimonials meglumine antimoniate (Glucantime) and 65% to 100% have been reported with combination therapy,
with relapse rates of around 10%.93,97 Dogs that relapse can
be retreated with one or more cycles of meglumine. Relapses
can occur years after treatment is initiated, even in the face
of allopurinol treatment. Rarely, xanthine urolithiasis com-
plicates long-term allopurinol treatment.97 A liposomal form
of meglumine antimoniate (LMA) (6.5 mg/kg of stiboglu-
conate every 4 days), when used together with allopurinol
(20 mg/kg PO q24h) and administered for 140 days, signifi-
cantly reduced clinical signs and parasite load and blocked
transmission of the parasite to sandflies, but was associated
with apparent cure in only 50% of treated dogs.98 Unfortu-
nately, resistance to antimonials has emerged among some
Leishmania isolates in highly endemic areas such as southern
Europe. Antimonials are also not readily available for treat-
ment of dogs in the United States.
Miltefosine activates proteases in Leishmania spp. and
causes apoptotic death of the parasite (see Chapter 10). Milt-
efosine can be used as an alternative to meglumine antimoni-
ate in conjunction with allopurinol for 1 month, after which
monotherapy with allopurinol is continued.99 Unfortunately
FIGURE 74-7  Histopathology of the kidney in a dog with visceral leishmaniosis. this combination protocol does not result in parasitologic cure,
There is marked thickening of glomerular capillary loops (arrows) indicative of glomerulo- and relapse is common. Even a second cycle of miltefosine treat-
nephritis. H&E stain. Bar = 50 µm. (Courtesy Dr. Christine Petersen, Iowa State University.) ment failed to eliminate the parasite in one study.99

TABLE 74-3
Suggested Drug Doses for Treatment of Canine Visceral Leishmaniosis
Drug Dose (mg/kg) Route Interval (hours) Duration and Comments
Allopurinol 10 PO 12 At least 6 to 12 months or until both clinical signs
disappear and quantitative serology becomes
negative
Meglumine 75-100 SC 24 30 days. Use with allopurinol. Meglumine antimoniate
antimoniate 50 12 is the first-line treatment where available.
Miltefosine 2 PO 24 28 days. Use with allopurinol where miltefosine is
available.
Lipid-complexed 3 IV Every 48 hours on Dilute to 1 mg/mL in D5W and administer over
amphotericin B Monday, Wednesday, 1 to 2 hours until a cumulative dose of 21 mg/kg
and Friday is reached. Monitor kidney values (see Chapter
9). Optimal dosing schedule for dogs is unknown.
720 SECTION 4  Protozoal Diseases

TABLE 74-4
Clinical Staging of Canine Visceral Leishmaniosis Based on Serologic Status, Clinical Signs, Laboratory Findings,
Type of Treatment, and Prognosis for Each Stage
Clinical Laboratory
Stage Antibody Titer Clinical Signs Findings Therapy Prognosis
Stage I Negative or low Mild signs such as Usually none, creatinine None, allopurinol Good
positive (<3- to peripheral lymphade- <1.4 mg/dL, UPC <0.5 monotherapy,
4-fold increase above nopathy or papular or combination
the laboratory cutoff) dermatitis therapy
Stage II Positive (low or high) Diffuse or symmetrical Anemia, hyperglobulinemia, Combination Good to guarded
cutaneous lesions, and/or hypoalbuminemia therapy
ulcerations, anorexia, present.
weight loss, fever, Substage a: nonazotemic,
and/or epistaxis nonproteinuric
Substage b: nonazotemic,
UPC 0.5-1
Stage III Medium to high Stage II abnormalities Stage II abnormalities Combination Guarded to poor
plus signs of immune- plus IRIS* stage I with therapy
complex disease UPC >1, or IRIS stage II
(vasculitis, arthritis,
glomerulonephritis)
Stage IV Medium to high Stage III abnormali- Stage II abnormalities Consider mono- Poor
ties plus nephrotic plus IRIS stage III or IV; therapy with al-
syndrome, pulmonary marked proteinuria lopurinol. Follow
thromboembolism, (UPC >5) IRIS guidelines
or end-stage renal for treatment of
disease chronic kidney
disease.

Data from Solano-Gallego L, Koutinas A, Miro G, et al. Directions for the diagnosis, clinical staging, treatment and prevention of canine leishmaniosis.
Vet Parasitol 2009;165:1-18.
*International Renal Interest Society, http://www.iris-kidney.com/guidelines/en

Lipid-complexed amphotericin B is the treatment of choice complications of uremia and/or protein-losing nephropathy,
for human visceral leishmaniasis. Limited reports of the use such as antacids, antiemetics, phosphate binders, low-protein
of both deoxycholate and lipid-complexed forms of ampho- diets, and antihypertensive drugs.
tericin B to treat dogs with leishmaniosis exist.100,101 The
need for intravenous administration of amphotericin B and Immunity and Vaccination
its nephrotoxicity has limited its use for treatment of dis-
ease in dogs. In one study, use of liposomal amphotericin B Although immunity to leishmaniosis is complex and not well
(Ambisome) to treat canine leishmaniosis was associated understood, it is clear that it is strongly dependent on the develop-
with rapid resolution of clinical signs, but cytologic evidence ment of a robust Th1-cell–mediated immune response. Production
of parasites persisted in lymph node aspirates.100 However, of cytokines such as IFN-γ and TNF-α activates macrophages to
treatment was administered for less than 2 weeks (and for destroy amastigotes, which occurs through nitric oxide–­mediated
some groups of dogs in the study, only 3 days). The FDA- mechanisms. Dogs that resist leishmaniosis exhibit strong delayed
approved regimen for liposomal amphotericin B treatment type hypersensitivity responses to leishmanial antigen when it is
of immunocompetent human patients consists of seven infu- administered intradermally. A balance between Th1 and Th2
sions over 21 days (days 1 through 5, 10, and 21), for a total immune responses appears to be important in determining the
dose of 21 mg/kg.102 clinical outcome of infection.
Because of the likelihood of relapse in dogs after treatment, the Several vaccines against leishmaniosis have been evaluated in
World Health Organization does not advocate treatment of pet dogs under field conditions. One of these vaccines, Leishmune,
dogs with anti-Leishmania chemotherapy protocols used to treat became licensed for prevention of leishmaniasis in dogs in Brazil
humans and instead suggests monotherapy with allopurinol.103 in 2011.53 Leishmune is composed of a promastigote antigen
(fucose-mannose ligand) and the adjuvant saponin.104,105 The
Supportive Care vaccine has not been approved in Europe because of adverse
Dogs with renal failure secondary to Leishmania infection may effects induced by the saponin adjuvant. Efficacy in initial
require supplemental fluid therapy and medications that address field trials was approximately 80% and has correlated with a
CHAPTER 74  Leishmaniosis 721

reduction in human leishmaniasis in the area.53 The vaccine is associated with reduction in human cases in the area. Nev-
also licensed in Brazil for immunotherapy, and reduces clinical ertheless, other canine species, such as foxes, may also play
signs in infected dogs,106 but when used without antiprotozoal a role as reservoirs.
drugs did not reduce 4.5-year mortality when compared with Human CL occurs in the Americas, the Mediterranean
infected control dogs.107 Another vaccine (CaniLeish, Virbac) basin, and western Asia from the Middle East to Central Asia,
composed of purified excreted-secreted proteins of Leishmania and especially in Afghanistan, Algeria, Colombia, Brazil, Iran,
infantum (LiESP) is licensed in Europe for prevention of canine Syria, Ethiopia, North Sudan, Costa Rica, and Peru. In Africa
leishmaniosis. This vaccine stimulates a Th1-dominated anti–L. and western Asia, CL is caused by L. major, L. tropica, and
infantum response within 3 weeks of administration.108 L. aethiopica, whereas in the Americas, the most common
causes are L. mexicana and L. braziliensis. Mucocutaneous
Prevention disease occurs primarily in South America and is caused by
L. braziliensis, for which rodents and other small mammals are
In addition to vaccination, use of ectoparasiticides such as the primary reservoirs.117
­deltamethrin-impregnated dog collars or spot-on repellants Recommendations of the World Health organization for
that contain permethrin can prevent canine leishmaniosis.111-114 the control of leishmaniasis in humans consist of treatment of
Indoor housing or use of fine-mesh netting around dogs and human disease; culling of wild and feral seropositive dogs; and
humans when sandflies feed at night (from dusk to dawn) is also insecticidal treatment of human homes, such as with pyrethroid
recommended. Because of transplacental transmission, neuter- sprays.103 The effectiveness of the culling of seropositive or
ing may also reduce the spread of Leishmania.6 Potential blood infected dogs is controversial.42,118 At present this is only under-
donors should be screened with serology and PCR assays and taken systematically in Brazil. Concerns raised include the ethics
should only be used if both assays are negative.29 of destruction of pet dogs, delays between detection of seroposi-
A dopamine-based oral suspension Leisguard is available in tive dogs and culling, and the persistence of other subclinically or
some parts of Europe for prevention of leishmaniosis in dogs clinically infected canine and wildlife reservoirs. Vaccination of
that travel to regions where the disease is endemic. This con- dogs may be more effective than the culling of seropositive dogs
tains the dopamine D2 receptor antagonist domperidone, which and could overcome the ethical concerns that relate to culling.53
activates phagocytic cells and enhances intracellular killing
of the parasite.109 A prospective, randomized controlled trial FELINE LEISHMANIOSIS
showed that Leisguard prevented transmission of leishmaniosis
to naïve dogs in a kennel.115 The drug was administered q24h Cats are relatively resistant to leishmaniosis, but cases have
for two 30-day periods at the estimated beginning and end of been described from southern Europe, the Middle East,
the vector activity period. Leisguard may also hasten recovery Latin America, and Texas.119,120 The cat from Texas was
when used for treatment of affected dogs in combination with infected with L. mexicana. L. infantum and a variety of other
meglumine antimoniate and allopurinol,110 but additional stud- Leishmania species such as L. braziliensis and L. venezu-
ies are required. elensis have also been isolated from cats. Serologic studies
of cats in endemic areas have generally revealed seropreva-
Public Health Aspects lences of less than 10%, although 34 (36%) of 95 stray cats
from the Yucatán peninsula in Mexico had antibodies to
An estimated 12 million people worldwide have leishmani- L. infantum, L. braziliensis, or L. mexicana.119 Disease in cats
asis, and at least 350 million people are estimated to be at risk. may be associated with concurrent FIV infection.121,122 The
Approximately 0.2 to 0.4 million new cases of VL and 0.7 to extent to which cats are a reservoir for infection of humans
1.2 million cases of CL are believed to occur in humans annu- is unclear.
ally, and it has been estimated that 20,000 to 40,000 leishmani- Clinical signs of leishmaniosis in cats include cutaneous
asis deaths occur per year.116 lesions, weight loss, lymphadenopathy, oral ulceration, muco-
VL in humans is also known as kala-azar. It is caused mostly purulent ocular and nasal discharge, keratitis, altered mentation,
by L. donovani and L. infantum (referred to as L. chagasi in Cen- diarrhea, and/or tachypnea. Cutaneous lesions consist of nodules,
tral and South America), but also by L. tropica. More than scaling and crusting, erythema, ulceration, and alopecia. Cutane-
90% of VL cases globally occur in India, Bangladesh, Sudan, ous lesions can occur anywhere and may be localized or general-
South Sudan, Brazil, and Ethiopia. Many cases are due to L. ized, but are often on the pinnae, bridge of the nose, limbs, or
donovani, for which humans are the primary reservoir.116 dorsal trunk. In one study, 50% of cats with skin lesions had
L. infantum causes VL in central Asia, China, the Mediter- evidence of visceral spread.121
ranean basin, the Middle East, and Latin America. Most dis- Diagnosis of leishmaniosis in cats is similar to that in dogs,
ease associated with L. infantum infection occurs in children although cats often have low antibody titers.121 Limited infor-
(infantile leishmaniasis) and immunocompromised adults, mation exists in regard to treatment of feline leishmaniosis.
such as those with AIDS. L. infantum is also responsible for Treatment with allopurinol monotherapy may result in clinical
some CL in humans. Dogs are considered the primary reser- improvement or disease resolution in some cats.123 Surgery may
voirs for L. infantum, and control of disease in dogs has been be curative for localized cutaneous lesions.12
722 SECTION 4  Protozoal Diseases

CASE EXAMPLE Laboratory Findings:


CBC:
HCT 32.4% (40%-55%)
Signalment: “Fausto,” an 8-year-old male neutered husky mix MCV 62.7 fL (65-75 fL)
from northern California MCHC 35.2 g/dL (33-36 g/dL)
History: Fausto was referred to the University of California, Davis, Reticulocyte count 54,500 cells/µL
Veterinary Medical Teaching Hospital (VMTH) for evaluation WBC 9300 cells/µL (6000-13,000 cells/µL)
of suspected autoimmune disease. The dog had a 6-month Neutrophils 7533 cells/µL (3000-10,500 cells/µL)
history of progressive skin lesions that began as crusting and Lymphocytes 930 cells/µL (1000-4000 cells/µL)
ulceration around the elbow and stifle joints. This was followed Monocytes 651 cells/µL (150-1200 cells/µL)
by development of similar lesions above both eyes and on Eosinophils 186 cells/µL (0-1500 cells/µL)
the dog’s ears and dorsal muzzle. Three months after the skin Platelets 338,000 platelets/µL (150,000-400,000 platelets/µL).
lesions appeared, Fausto became stiff and lethargic and was Serum Chemistry Profile:
lame and reluctant to walk. At that time, Fausto was taken to Sodium 140 mmol/L (145-154 mmol/L)
a local veterinary clinic where an injection of triamcinolone Potassium 4.4 mmol/L (4.1-5.3 mmol/L)
acetonide (Vetalog) was administered, and he was prescribed Chloride 107 mmol/L (105-116 mmol/L)
carprofen and trimethoprim-sulfamethoxazole. There was Bicarbonate 15 mmol/L (16-26 mmol/L)
no improvement in the clinical signs. The day before he Phosphorus 3.8 mg/dL (3.0-6.2 mg/dL)
was brought to the VMTH, the owner had taken Fausto to a Calcium 10.4 mg/dL (9.9-11.4 mg/dL)
local emergency clinic because he was reluctant to move. BUN 16 mg/dL (8-31 mg/dL)
Blood work and arthrocentesis (left and right tarsal joints) Creatinine 0.9 mg/dL (0.3-1.2 mg/dL)
were performed. The blood work showed a normocytic, Glucose 109 mg/dL (70-118 mg/dL)
normochromic nonregenerative anemia (HCT 31%); mild Total protein 10.7 g/dL (5.4-7.4 g/dL)
hypoalbuminemia (2.4 g/dL); and moderate hyperglobulinemia Albumin 2.1 g/dL (2.9-4.2 g/dL)
(7.7 g/dL). Cytologic examination of the synovial fluid revealed Globulin 8.6 g/dL (2.3-4.4 g/dL)
large numbers of nondegenerate neutrophils, which was ALT 12 U/L (19-67 U/L)
considered consistent with immune-mediated polyarthritis. AST 39 U/L (21-54 U/L)
Cephalexin and carprofen were prescribed, and the dog was ALP 43 U/L (15-127 U/L)
referred for further evaluation. GGT 3 U/L (0-6 U/L)
The owner had not noticed any coughing, sneezing, vomit- Cholesterol 191 mg/dL (135-345 mg/dL)
ing, diarrhea, or changes in thirst or urination. Fausto was Total bilirubin 0.1 mg/dL (0-0.4 mg/dL).
fed 3 cups of a commercial dry chicken and rice–based Serum Protein Electrophoresis: There was a marked increase
dog food once a day, and his appetite was good. He in gamma-globulin with mild hypoalbuminemia. The
was fully vaccinated for rabies, canine distemper virus, gammopathy was polyclonal.
canine adenovirus, and canine parvovirus. The dog did Urinalysis (Cystocentesis): SGr 1.037; pH 7.0, 4+ protein, 1+
not receive heartworm prophylaxis medication but was bilirubin, 1+ hemoprotein, no glucose or ketones, rare WBC/
treated with fipronil once a month for flea control. There HPF, 1-3 RBC/HPF, rare hyaline casts, few lipid droplets.
was one other healthy dog in the household. The owner Urine Protein-to-Creatinine Ratio: 2.99 (normal, <0.5).
and his dogs had moved to California from Barcelona, Microbiologic Testing: IFA serology for antibodies for
Spain, a few weeks earlier, around the time that the skin Ehrlichia spp., Anaplasma spp., Rickettsia spp., Bartonella
lesions appeared. In Spain, Fausto had worn a collar to clarridgeiae, Bartonella vinsonii subsp. berkhoffii, and
prevent insect bites. Bartonella henselae: Negative.
Physical Examination: Western immunoblot for antibodies to Borrelia burgdorferi:
Body Weight: 34.4 kg. Negative.
General: Quiet, alert, and responsive, but reluctant to ambulate. Knott’s and ELISA antigen test for Dirofilaria immitis infection:
T = 102.7°F (39.3°C), HR = 132 beats/min, panting but eupneic, Negative.
mucous membranes moist and pink. Aerobic bacterial, anaerobic bacterial, and Mycoplasma
Integument: Multiple focal regions of crusting, erythema, culture of synovial fluid: Negative.
hyperpigmentation, thickening, and sometimes ulceration Aspiration cytology (right and left superficial cervical and popli-
were present over the elbows and stifles, prepuce, ventral teal lymph nodes): A heterogeneous population of lympho-
aspect of the mandible, above both eyes, and on both pinnae. cytes was present, which consisted primarily of small mature
Moderate pruritis was present. lymphocytes and slightly increased numbers of intermedi-
Musculoskeletal: BCS 5/9. The dog was able to walk but had ate-sized lymphocytes and lymphoblasts. Large numbers of
a shifting lameness. Pain was detected on extension of the plasma cells were noted. A few macrophages, nondegener-
joints, but there was no palpable effusion present. ate neutrophils, and eosinophils were scattered throughout
Abdominal Palpation: Splenomegaly was detected. the specimen.
Lymph Nodes: Superficial cervical and popliteal lymph nodes Bone marrow aspiration cytology: Many unit particles were pres-
were all enlarged (approximately 3 cm in diameter). ent. Unit particles were normocellular, and a small amount
All Other Systems: No significant abnormalities were detected. of storage iron was noted. There were normal numbers of
CHAPTER 74  Leishmaniosis 723

megakaryocytes. The myeloid-to-erythroid ratio was mildly hypoalbuminemia (1.9 g/dL). A drug reaction was suspected,
increased; both series were synchronous and complete. In- and the cephalexin was replaced with clavulanic acid–
creased numbers of plasma cells were noted throughout amoxicillin. Over the next month, the skin lesions improved.
the specimen, with few small lymphocytes. Macrophages The owner subsequently obtained meglumine antimoniate
occasionally contained intracytoplasmic organisms consis- from Spain, and treatment was changed to allopurinol (10
tent with Leishmania spp. amastigotes. Interpretation: Mild mg/kg PO q12h) and meglumine antimoniate (88 mg/kg SC
erythroid hypoplasia, Leishmania spp., and mild plasma cell q24h for 1 month). However, 6 days later, the dog developed
hyperplasia. prolapsed third eyelids and was unable to catch treats when
Skin biopsy: There was a thick band of inflammatory cells that they were thrown to him. Physical examination showed
extended from the superficial to the mid-dermis. The in- healing skin lesions with evidence of hair regrowth, prolapsed
flammatory infiltrate was composed of dense sheets of third eyelids, risus sardonicus, and erect ears. A presumptive
large foamy macrophages admixed with smaller numbers of diagnosis of localized tetanus was made. A serum chemistry
lymphocytes and plasma cells and few neutrophils. Many of panel showed an albumin of 2.5 g/dL and a globulin of 6.4 g/
the large macrophages contained multiple, round to oval, dL. The urine protein-to-creatinine ratio was 0.2. Fausto was
intracytoplasmic basophilic organisms that were 1 to 2 µm treated with metronidazole (10 mg/kg PO q8h for 14 days)
in diameter. The epidermis was hyperplastic and hyperkera- and tetanus antitoxin (20,000 U in 250 mL saline over
totic and was covered by exudate admixed with keratin, de- 1 hour, IV, once). Clinical signs of tetanus had almost resolved
generate neutrophils, and cellular debris. 11 days later when the owner was contacted by telephone.
Diagnosis: Canine visceral leishmaniosis. The dog was subsequently lost to follow-up.
Treatment and Outcome: Fausto was initially treated Comments: This case is an interesting example of travel-
with a combination of allopurinol (10 mg/kg PO q8h), related canine visceral leishmaniosis that was ultimately
ketoconazole (9 mg/kg PO q12h), cephalexin (30 mg/kg PO complicated by what appeared to be a cutaneous drug
q12h for 4 weeks, for secondary bacterial pyoderma), and reaction to cephalexin and then localized tetanus. The
carprofen (4.4 mg/kg PO q24h). At a recheck examination 2 diagnosis of leishmaniosis was suspected based on the
weeks later, the dog’s owner reported that 1 or 2 days after clinical signs together with the travel history from southern
treatment was started, Fausto became intensely pruritic. Europe. The bone marrow aspirate and skin biopsies were
Fausto’s appetite also decreased and he developed increased obtained simultaneously for the sole purpose of diagnosis
thirst and urination, but the lameness resolved. Physical of leishmaniosis; impression smears of the skin lesions failed
examination showed severe crusting and ulceration of to yield organisms, and the lymph node aspirates were also
the face, with an associated foul odor. This extended from negative. The owner and his dog traveled back to Spain in
the dorsal nasal planum to the temporal and supraocular order to obtain the meglumine antimoniate, which could
regions, as well as the inner aspect and tips of the pinnae. not be obtained in the United States. The development of
The lesions on the limbs were static or slightly improved from tetanus may have followed wound contamination or use
the last visit. A CBC was unchanged, and a serum chemistry of contaminated needles by the owner for subcutaneous
profile showed worsened hyperglobulinemia (9.3 g/dL) and injection of the meglumine.

SUGGESTED READINGS 4. Owens SD, Oakley DA, Marryott K, et al. Transmission of visceral
leishmaniasis through blood transfusions from infected English fox-
Maroli M, Gradoni L, Oliva G, et al. Guidelines for prevention of leish- hounds to anemic dogs. J Am Vet Med Assoc. 2001;219:1076-1083.
maniasis in dogs. J Am Vet Med Assoc. 2010;236:1200-1206. 5. Naucke TJ, Lorentz S. First report of venereal and vertical trans-
Oliva G, Roura X, Crotti A, et al. Guidelines for treatment of leishmaniasis mission of canine leishmaniosis from naturally infected dogs in
in dogs. J Am Vet Med Assoc. 2010;236:1192-1198. Germany. Parasit Vectors. 2012;5:67.
Paltrinieri S, Solano-Gallego L, Fondati A, et al. Guidelines for diagnosis 6. Boggiatto PM, Gibson-Corley KN, Metz K, et  al. Transplacental
and clinical classification of leishmaniasis in dogs. J Am Vet Med Assoc. transmission of Leishmania infantum as a means for continued dis-
2010;236:1184-1191. ease incidence in North America. PLoS Negl Trop Dis. 2011;5:e1019.
Solano-Gallego L, Koutinas A, Miro G, et al. Directions for the diagno- 7. Silva FL, Oliveira RG, Silva TM, et al. Venereal transmission of
sis, clinical staging, treatment and prevention of canine leishmaniosis. canine visceral leishmaniasis. Vet Parasitol. 2009;160:55-59.
Vet Parasitol. 2009;165:1-18. 8. Leblois R, Kuhls K, Francois O, et al. Guns, germs and dogs: on
Solano-Gallego L, Miro G, Koutinas A, et  al. LeishVet guidelines for the origin of Leishmania chagasi. Infect Genet Evol. 2011;11:
the practical management of canine leishmaniosis. Parasit Vectors. 1091-1095.
2011;4:86. 9. Sykes JE. Unpublished observations, 2012.
10. Teske E, van Knapen F, Beijer EG, et  al. Risk of infection with
REFERENCES Leishmania spp. in the canine population in the Netherlands. Acta
1. Dutta AK. Pursuit of medical knowledge: Charles Donovan Vet Scand. 2002;43:195-201.
(1863-1951) on kala-azar in India. J Med Biogr. 2008;16:72-76. 11. Shaw SE, Langton DA, Hillman TJ. Canine leishmaniosis in the
2. Schonian G, Mauricio I, Cupolillo E. Is it time to revise the United Kingdom: a zoonotic disease waiting for a vector? Vet
nomenclature of Leishmania? Trends Parasitol. 2010;26:466-469. Parasitol. 2009;163:281-285.
3. Antinori S, Schifanella L, Corbellino M. Leishmaniasis: new 12. Rufenacht S, Sager H, Muller N, et al. Two cases of feline leish-
insights from an old and neglected disease. Eur J Clin Microbiol maniosis in Switzerland. Vet Rec. 2005;156:542-545.
Infect Dis. 2012;31:109-118.
724 SECTION 4  Protozoal Diseases

13. Kawamura Y, Yoshikawa I, Katakura K. Imported leishmaniasis 34. Cardoso L, Yisaschar-Mekuzas Y, Rodrigues FT, et  al. Canine
in dogs, U.S. military bases. Japan. Emerg Infect Dis. 2010;16: babesiosis in northern Portugal and molecular characteriza-
2017-2019. tion of vector-borne co-infections. Parasit Vectors. 2010;3:
14. Moreno J, Alvar J. Canine leishmaniasis: epidemiological risk and 27.
the experimental model. Trends Parasitol. 2002;18:399-405. 35. Mekuzas Y, Gradoni L, Oliva G, et al. Ehrlichia canis and Leish-
15. Berrahal F, Mary C, Roze M, et al. Canine leishmaniasis: identifi- mania infantum co-infection: a 3-year longitudinal study in nat-
cation of asymptomatic carriers by polymerase chain reaction and urally exposed dogs. Clin Microbiol Infect. 2009;15(suppl 2):
immunoblotting. Am J Trop Med Hyg. 1996;55:273-277. 30-31.
16. Solano-Gallego L, Morell P, Arboix M, et al. Prevalence of Leish- 36. Tabar MD, Francino O, Altet L, et  al. PCR survey of vector-
mania infantum infection in dogs living in an area of canine leish- borne pathogens in dogs living in and around Barcelona, an area
maniasis endemicity using PCR on several tissues and serology. endemic for leishmaniasis. Vet Rec. 2009;164:112-116.
J Clin Microbiol. 2001;39:560-563. 37. Abreu-Silva AL, Calabrese KS, Cupolilo SM, et  al. Histopatho-
17. Leontides LS, Saridomichelakis MN, Billinis C, et  al. A cross- logical studies of visceralized Leishmania (Leishmania) amazo-
sectional study of Leishmania spp. infection in clinically healthy nensis in mice experimentally infected. Vet Parasitol. 2004;121:
dogs with polymerase chain reaction and serology in Greece. Vet 179-187.
Parasitol. 2002;109:19-27. 38. Guessous-Idrissi N, Berrag B, Riyad M, et al. Short report: Leish-
18. Tanczos B, Balogh N, Kiraly L, et al. First record of autochtho- mania tropica: etiologic agent of a case of canine visceral leish-
nous canine leishmaniasis in Hungary. Vector Borne Zoonotic Dis. maniasis in northern Morocco. Am J Trop Med Hyg. 1997;57:
2012;12:588-594. 172-173.
19. Ready PD. Leishmaniasis emergence in Europe. Euro Surveill. 39. Hassan MM, Osman OF, El-Raba’a FM, et al. Role of the domestic
2010;15:19505. dog as a reservoir host of Leishmania donovani in eastern Sudan.
20. Shang LM, Peng WP, Jin HT, et  al. The prevalence of canine Parasit Vectors. 2009;2:26.
Leishmania infantum infection in Sichuan Province, southwest- 40. Reithinger R, Davies CR. American cutaneous leishmaniasis in
ern China detected by real time PCR. Parasit Vectors. 2011;4: domestic dogs: an example of the use of the polymerase chain
173. reaction for mass screening in epidemiological studies. Trans R
21. Cortes S, Vaz Y, Neves R, et  al. Risk factors for canine leish- Soc Trop Med Hyg. 2002;96(suppl 1):S123-S126.
maniasis in an endemic Mediterranean region. Vet Parasitol. 41. Marco JD, Barroso PA, Calvopina M, et  al. Species assignation
2012;189:189-196. of Leishmania from human and canine American tegumentary
22. Cardoso L, Rodrigues M, Santos H, et  al. Sero-epidemiological leishmaniasis cases by multilocus enzyme electrophoresis in North
study of canine Leishmania spp. infection in the municipality of Argentina. Am J Trop Med Hyg. 2005;72:606-611.
Alijo (Alto Douro, Portugal). Vet Parasitol. 2004;121:21-32. 42. Silva DA, Madeira MF, Teixeira AC, et al. Laboratory tests per-
23. Galvez R, Miro G, Descalzo MA, et  al. Emerging trends in the formed on Leishmania seroreactive dogs euthanized by the leish-
seroprevalence of canine leishmaniasis in the Madrid region maniasis control program. Vet Parasitol. 2011;179:257-261.
(central Spain). Vet Parasitol. 2010;169:327-334. 43. Velez ID, Carrillo LM, Lopez L, et al. An epidemic outbreak of
24. Abranches P, Silva-Pereira MC, Conceicao-Silva FM, et al. Canine canine cutaneous leishmaniasis in Colombia caused by Leishma-
leishmaniasis: pathological and ecological factors influencing nia braziliensis and Leishmania panamensis. Am J Trop Med Hyg.
transmission of infection. J Parasitol. 1991;77:557-561. 2012;86:807-811.
25. Bigeli JG, Oliveira Jr WP, Teles NM. Diagnosis of Leishma- 44. Tolezano JE, Uliana SR, Taniguchi HH, et  al. The first records
nia (Leishmania) chagasi infection in dogs and the relationship of Leishmania (Leishmania) amazonensis in dogs (Canis famil-
with environmental and sanitary aspects in the municipality iaris) diagnosed clinically as having canine visceral leishmaniasis
of Palmas, State of Tocantins, Brazil. Rev Soc Bras Med Trop. from Aracatuba County, Sao Paulo State, Brazil. Vet Parasitol.
2012;45:18-23. 2007;149:280-284.
26. Solano-Gallego L, Llull J, Ramos G, et al. The Ibizian hound pres- 45. Sun K, Guan W, Zhang JG, et al. Prevalence of canine leishmania-
ents a predominantly cellular immune response against natural sis in Beichuan County, Sichuan, China and phylogenetic evidence
Leishmania infection. Vet Parasitol. 2000;90:37-45. for an undescribed Leishmania sp. in China based on 7SL RNA.
27. Miranda S, Roura X, Picado A, et al. Characterization of sex, age, Parasit Vectors. 2012;5:75.
and breed for a population of canine leishmaniosis diseased dogs. 46. Freeman KS, Miller MD, Breitschwerdt EB, et al. Leishmaniasis in a
Res Vet Sci. 2008;85:35-38. dog native to Colorado. J Am Vet Med Assoc. 2010;237:1288-1291.
28. Quilez J, Martinez V, Woolliams JA, et  al. Genetic control of 47. Anderson DC, Buckner RG, Glenn BL, et al. Endemic canine leish-
canine leishmaniasis: genome-wide association study and genomic maniasis. Vet Pathol. 1980;17:94-96.
selection analysis. PLoS One. 2012;7:e35349. 48. Sellon RK, Menard MM, Meuten DJ, et al. Endemic visceral leish-
29. Solano-Gallego L, Koutinas A, Miro G, et al. Directions for the maniasis in a dog from Texas. J Vet Intern Med. 1993;7:16-19.
diagnosis, clinical staging, treatment and prevention of canine 49. Swenson CL, Silverman J, Stromberg PC, et al. Visceral leishmani-
leishmaniosis. Vet Parasitol. 2009;165:1-18. asis in an English foxhound from an Ohio research colony. J Am
30. Marino G, Gaglio G, Zanghi A. Clinicopathological study of canine Vet Med Assoc. 1988;193:1089-1092.
transmissible venereal tumour in leishmaniotic dogs. J Small Anim 50. Gaskin AA, Schantz P, Jackson J, et al. Visceral leishmaniasis in a
Pract. 2012;53:323-327. New York foxhound kennel. J Vet Intern Med. 2002;16:34-44.
31. Mozos E, Perez J, Day MJ, et al. Leishmaniosis and generalized 51. Freeman K. Update on the diagnosis and management of Leish-
demodicosis in three dogs: a clinicopathological and immunohis- mania spp. infections in dogs in the United States. Top Companion
tochemical study. J Comp Pathol. 1999;120:257-268. Anim Med. 2010;25:149-154.
32. Ciaramella P, Oliva G, Luna RD, et  al. A retrospective clinical 52. Molina R, Amela C, Nieto J, et  al. Infectivity of dogs naturally
study of canine leishmaniasis in 150 dogs naturally infected by infected with Leishmania infantum to colonized Phlebotomus per-
Leishmania infantum. Vet Rec. 1997;141:539-543. niciosus. Trans R Soc Trop Med Hyg. 1994;88:491-493.
33. Cardoso L, Mendao C, Madeira de Carvalho L. Prevalence of 53. Palatnik-de-Sousa CB. Vaccines for canine leishmaniasis. Front
Dirofilaria immitis, Ehrlichia canis, Borrelia burgdorferi sensu lato, Immunol. 2012;3:69.
Anaplasma spp. and Leishmania infantum in apparently healthy 54. Altet L, Francino O, Solano-Gallego L, et al. Mapping and sequenc-
and CVBD-suspect dogs in Portugal—a national serological study. ing of the canine NRAMP1 gene and identification of mutations in
Parasit Vectors. 2012;5:62. leishmaniasis-susceptible dogs. Infect Immun. 2002;70:2763-2771.
CHAPTER 74  Leishmaniosis 725

55. Sanchez-Robert E, Altet L, Sanchez A, et  al. Polymorphism of 78. Font A, Closa JM, Mascort J. Monoclonal gammopathy in a dog
Slc11a1 (Nramp1) gene and canine leishmaniasis in a case-control with visceral leishmaniasis. J Vet Intern Med. 1994;8:233-235.
study. J Hered. 2005;96:755-758. 79. Foglia Manzillo V, Restucci B, Pagano A, et  al. Pathological
56. Quinnell RJ, Kennedy LJ, Barnes A, et al. Susceptibility to visceral changes in the bone marrow of dogs with leishmaniosis. Vet Rec.
leishmaniasis in the domestic dog is associated with MHC class II 2006;158:690-694.
polymorphism. Immunogenetics. 2003;55:23-28. 80. Tropia de Abreu R, Carvalho MG, Carneiro CM, et al. Influence
57. Petanides TA, Koutinas AF, Mylonakis ME, et al. Factors asso- of clinical status and parasite load on erythropoiesis and leuco-
ciated with the occurrence of epistaxis in natural canine leish- poiesis in dogs naturally infected with Leishmania (Leishmania)
maniasis (Leishmania infantum). J Vet Intern Med. 2008;22: chagasi. PLoS One. 2011;6:e18873.
866-872. 81. Giudice E, Passantino A. Detection of Leishmania amastigotes in
58. Paciello O, Oliva G, Gradoni L, et al. Canine inflammatory myop- peripheral blood from four dogs—short communication. Acta Vet
athy associated with Leishmania infantum infection. Neuromus- Hung. 2011;59:205-213.
cul Disord. 2009;19:124-130. 82. Galvez R, Miro G, Descalzo MA, et al. Questionnaire-based sur-
59. Lopez R, Lucena R, Novales M, et al. Circulating immune com- vey on the clinical management of canine leishmaniosis in the
plexes and renal function in canine leishmaniasis. Zentralbl Vet- Madrid region (central Spain). Prev Vet Med. 2011;102:59-65.
erinarmed B. 1996;43:469-474. 83. Marcondes M, Biondo AW, Gomes AA, et  al. Validation of a
60. Terrazzano G, Cortese L, Piantedosi D, et al. Presence of anti-plate- Leishmania infantum ELISA rapid test for serological diagnosis of
let IgM and IgG antibodies in dogs naturally infected by Leishma- Leishmania chagasi in dogs. Vet Parasitol. 2011;175:15-19.
nia infantum. Vet Immunol Immunopathol. 2006;110:331-337. 84. de Amorim IF, Freitas E, Alves CF, et al. Humoral immunologi-
61. Smith BE, Tompkins MB, Breitschwerdt EB. Antinuclear anti- cal profile and parasitological statuses of Leishmune vaccinated
bodies can be detected in dog sera reactive to Bartonella vinsonii and visceral leishmaniasis infected dogs from an endemic area. Vet
subsp. berkhoffii, Ehrlichia canis, or Leishmania infantum anti- Parasitol. 2010;173:55-63.
gens. J Vet Intern Med. 2004;18:47-51. 85. Lombardo G, Pennisi MG, Lupo T, et al. Detection of Leishmania
62. Ginel PJ, Camacho S, Lucena R. Anti-histone antibodies in dogs infantum DNA by real-time PCR in canine oral and conjuncti-
with leishmaniasis and glomerulonephritis. Res Vet Sci. 2008;85: val swabs and comparison with other diagnostic techniques. Vet
510-514. Parasitol. 2012;184:10-17.
63. Slappendel RJ. Canine leishmaniasis. A review based on 95 cases 86. Martinez V, Quilez J, Sanchez A, et al. Canine leishmaniasis: the key
in The Netherlands. Vet Q. 1988;10:1-16. points for qPCR result interpretation. Parasit Vectors. 2011;4:57.
64. Naranjo C, Fondevila D, Altet L, et al. Evaluation of the presence 87. de Queiroz NM, da Silveira RC, de Noronha Jr AC, et al. Detec-
of Leishmania spp. by real-time PCR in the lacrimal glands of tion of Leishmania (L.) chagasi in canine skin. Vet Parasitol.
dogs with leishmaniosis. Vet J. 2012;193:168-173. 2011;178:1-8.
65. Mir F, Fontaine E, Reyes-Gomez E, et al. Subclinical leishmaniasis 88. Koutinas AF, Carlotti DN, Koutinas C, et al. Claw histopathology
associated with infertility and chronic prostatitis in a dog. J Small and parasitic load in natural cases of canine leishmaniosis associ-
Anim Pract. 2012;53:419-422. ated with Leishmania infantum. Vet Dermatol. 2010;21:572-577.
66. Font A, Roura X, Fondevila D, et al. Canine mucosal leishmani- 89. Costa FA, Goto H, Saldanha LC, et al. Histopathologic patterns
asis. J Am Anim Hosp Assoc. 1996;32:131-137. of nephropathy in naturally acquired canine visceral leishmani-
67. Viegas C, Requicha J, Albuquerque C, et  al. Tongue nodules in asis. Vet Pathol. 2003;40:677-684.
canine leishmaniosis—a case report. Parasit Vectors. 2012;5:120. 90. Nieto CG, Navarrete I, Habela MA, et al. Pathological changes in
68. Carrasco L, de Lara FC, Martin E, et al. Acute haemorrhagic pan- kidneys of dogs with natural Leishmania infection. Vet Parasitol.
creatitis associated with canine visceral leishmaniasis. Vet Rec. 1992;45:33-47.
1997;141:519-521. 91. Zatelli A, Borgarelli M, Santilli R, et al. Glomerular lesions in dogs
69. Pinto AJ, Figueiredo MM, Silva FL, et al. Histopathological and infected with Leishmania organisms. Am J Vet Res. 2003;64:558-561.
parasitological study of the gastrointestinal tract of dogs naturally 92. Dinhopl N, Mostegl MM, Richter B, et al. In situ hybridisation
infected with Leishmania infantum. Acta Vet Scand. 2011;53:67. for the detection of Leishmania species in paraffin wax-embedded
70. Toplu N, Aydogan A. An immunohistochemical study in cases canine tissues using a digoxigenin-labelled oligonucleotide probe.
with usual and unusual clinicopathological findings of canine vis- Vet Rec. 2011;169:525.
ceral leishmaniosis. Parasitol Res. 2011;109:1051-1057. 93. Noli C, Auxilia ST. Treatment of canine Old World visceral leish-
71. Lopez-Pena M, Aleman N, Munoz F, et al. Visceral leishmaniasis maniasis: a systematic review. Vet Dermatol. 2005;16:213-232.
with cardiac involvement in a dog: a case report. Acta Vet Scand. 94. Andrade HM, Toledo VP, Pinheiro MB, et  al. Evaluation of
2009;51:20. miltefosine for the treatment of dogs naturally infected with
72. Agut A, Corzo N, Murciano J, et  al. Clinical and radiographic L. infantum (= L. chagasi) in Brazil. Vet Parasitol. 2011;181:83-90.
study of bone and joint lesions in 26 dogs with leishmaniasis. Vet 95. Oliva G, Roura X, Crotti A, et  al. Guidelines for treatment of
Rec. 2003;153:648-652. leishmaniasis in dogs. J Am Vet Med Assoc. 2010;236:1192-1198.
73. de Souza AI, Juliano RS, Gomes TS, et al. Osteolytic osteomyeli- 96. Paradies P, Sasanelli M, Amato ME, et  al. Monitoring the
tis associated with visceral leishmaniasis in a dog. Vet Parasitol. reverse to normal of clinico-pathological findings and the disease
2005;129:51-54. free interval time using four different treatment protocols for
74. Vinuelas J, Garcia-Alonso M, Ferrando L, et al. Meningeal leish- canine leishmaniosis in an endemic area. Res Vet Sci. 2012;93:
maniosis induced by Leishmania infantum in naturally infected 843-847.
dogs. Vet Parasitol. 2001;101:23-27. 97. Torres M, Bardagi M, Roura X, et al. Long term follow-up of dogs
75. Cauduro A, Favole P, Lorenzo V, et al. Paraparesis caused by ver- diagnosed with leishmaniosis (clinical stage II) and treated with
tebral canal leishmaniotic granuloma in a dog. J Vet Intern Med. meglumine antimoniate and allopurinol. Vet J. 2011;188:346-351.
2011;25:398-399. 98. da Silva SM, Amorim IF, Ribeiro RR, et al. Efficacy of combined
76. Cavalcanti A, Lobo R, Cupolillo E, et al. Canine cutaneous leish- therapy with liposome-encapsulated meglumine antimoniate and
maniasis caused by neotropical Leishmania infantum despite of allopurinol in treatment of canine visceral leishmaniasis. Antimi-
systemic disease: a case report. Parasitol Int. 2012;61:738-740. crob Agents Chemother. 2012;56:2858-2867.
77. Freitas JC, Nunes-Pinheiro DC, Lopes Neto BE, et al. Clinical and 99. Manna L, Vitale F, Reale S, et al. Study of efficacy of miltefosine and
laboratory alterations in dogs naturally infected by Leishmania allopurinol in dogs with leishmaniosis. Vet J. 2009;182:441-445.
chagasi. Rev Soc Bras Med Trop. 2012;45:24-29.
726 SECTION 4  Protozoal Diseases

100. Oliva G, Gradoni L, Ciaramella P, et  al. Activity of liposomal leishmaniasis: effect on disease prevalence and the vectors (Diptera:
amphotericin B (AmBisome) in dogs naturally infected with Leish- Psychodidae) in a hyperendemic area. Vet Ther. 2002;3:485-492.
mania infantum. J Antimicrob Chemother. 1995;36:1013-1019. 113. Reithinger R, Coleman PG, Alexander B, et  al. Are insecticide-
101. Cortadellas O. Initial and long-term efficacy of a lipid emulsion of impregnated dog collars a feasible alternative to dog culling as a
amphotericin B desoxycholate in the management of canine leish- strategy for controlling canine visceral leishmaniasis in Brazil? Int
maniasis. J Vet Intern Med. 2003;17:808-812. J Parasitol. 2004;34:55-62.
102. Murray HW. Leishmaniasis in the United States: treatment in 114. Otranto D, Paradies P, Lia RP, et al. Efficacy of a combination of
2012. Am J Trop Med Hyg. 2012;86:434-440. 10% imidacloprid/50% permethrin for the prevention of leish-
103. Control of the leishmaniasis. Report of a meeting of the WHO maniasis in kennelled dogs in an endemic area. Vet Parasitol.
Expert Committee on the Control of Leishmaniases. Geneva, 22– 2007;144:270-278.
26 March 2010. http://www.who.int/leishmaniasis/surveillance/ 115. Gomez-Ochoa P, Sabate D, Homedes J, et al. Clinical efficacy of a
en/. Last accessed January 13, 2013. Leisguard-based program strategically established for the preven-
104. Parra LE, Borja-Cabrera GP, Santos FN, et al. Safety trial using tion of canine leishmaniosis in endemic areas with low prevalence
the Leishmune vaccine against canine visceral leishmaniasis in [Abstract]. In: 73rd Congresso Internazionale Multisala SCIVAC.
Brazil. Vaccine. 2007;25:2180-2186. 2012:545.
105. Nogueira FS, Moreira MA, Borja-Cabrera GP, et  al. Leishmune 116. Alvar J, Velez ID, Bern C, et  al. Leishmaniasis worldwide and
vaccine blocks the transmission of canine visceral leishmaniasis: global estimates of its incidence. PLoS One. 2012;7:e35671.
absence of Leishmania parasites in blood, skin and lymph nodes 117. Dantas-Torres F. Dogs as reservoirs for Leishmania braziliensis.
of vaccinated exposed dogs. Vaccine. 2005;23:4805-4810. Emerg Infect Dis. 2011;17:326-327:author reply 327.
106. Santos FN, Borja-Cabrera GP, Miyashiro LM, et al. Immunotherapy 118. Costa CH. How effective is dog culling in controlling zoonotic
against experimental canine visceral leishmaniasis with the saponin visceral leishmaniasis? A critical evaluation of the science, politics
enriched-Leishmune vaccine. Vaccine. 2007;25:6176-6190. and ethics behind this public health policy. Rev Soc Bras Med
107. Borja-Cabrera GP, Santos FN, Santos FB, et al. Immunotherapy Trop. 2011;44:232-242.
with the saponin enriched-Leishmune vaccine versus immuno- 119. Longoni SS, Lopez-Cespedes A, Sanchez-Moreno M, et al. Detec-
chemotherapy in dogs with natural canine visceral leishmaniasis. tion of different Leishmania spp. and Trypanosoma cruzi anti-
Vaccine. 2010;28:597-603. bodies in cats from the Yucatan Peninsula (Mexico) using an iron
108. Moreno J, Vouldoukis I, Martin V, et al. Use of a LiESP/QA-21 superoxide dismutase excreted as antigen. Comp Immunol Micro-
vaccine (CaniLeish) stimulates an appropriate Th1-dominated biol Infect Dis. 2012;35:469-476.
cell-mediated immune response in dogs. PLoS Negl Trop Dis. 120. Craig TM, Barton CL, Mercer SH, et al. Dermal leishmaniasis in
2012;6:e1683. a Texas cat. Am J Trop Med Hyg. 1986;35:1100-1102.
109. Gomez-Ochoa P, Sabate D, Homedes J, et  al. Use of the nitro- 121. Vides JP, Schwardt TF, Sobrinho LS, et  al. Leishmania chagasi
blue tetrazolium reduction test for the evaluation of Domperidone infection in cats with dermatologic lesions from an endemic area of vis-
effects on the neutrophilic function of healthy dogs. Vet Immunol ceral leishmaniosis in Brazil. Vet Parasitol. 2011;178:22-28.
Immunopathol. 2012;146:97-99. 122. Sobrinho LS, Rossi CN, Vides JP, et al. Coinfection of Leishma-
110. Lanaro E. Efficacy of a combined therapy with meglumine anti- nia chagasi with Toxoplasma gondii, feline immunodeficiency
moniate and domperidone for treatment of canine leishmaniosis. virus (FIV) and feline leukemia virus (FeLV) in cats from an
In: 73rd Congresso Internazionale Multisala SCIVAC. 2012:531. endemic area of zoonotic visceral leishmaniasis. Vet Parasitol.
111. Gavgani AS, Hodjati MH, Mohite H, et al. Effect of insecticide- 2012;187:302-306.
impregnated dog collars on incidence of zoonotic visceral leish- 123. Leiva M, Lloret A, Pena T, et al. Therapy of ocular and visceral
maniasis in Iranian children: a matched-cluster randomised trial. leishmaniasis in a cat. Vet Ophthalmol. 2005;8:71-75.
Lancet. 2002;360:374-379.
112. Giffoni JH, de Almeida CE, dos Santos SO, et  al. Evaluation
of 65% permethrin spot-on for prevention of canine visceral
CHAPTER 75

Babesiosis
Adam J. Birkenheuer

transmission between dogs, through fighting (and exchange of


Overview of Babesiosis in Dogs and Cats blood), or congenital transplacental transmission, is believed to
First Described: Early descriptions of intraerythrocytic para- be the most common route of transmission when competent tick
sites in dogs with signs of babesiosis were made in Africa vectors are absent.3-5
in 1893 (Hutcheon).1 The first documented case of canine When ticks are involved in transmission, sporozoites are
babesiosis in the United States was in 1934 (Eaton).2 released from the tick salivary glands as the tick feeds and they
Cause: Babesia canis, Babesia gibsoni, Babesia conradae (dogs); enter the bloodstream of the vertebrate host (Figure 75-1). They
Babesia felis, Babesia cati (cats), several other unnamed then attach to, and are endocytosed by, erythrocytes. Within
species (phylum Apicomplexa) erythrocytes, they undergo asexual reproduction (merogony),
and the daughter cells infect new erythrocytes. A naïve tick then
Affected Hosts: Domestic dogs and cats
ingests infected erythrocytes. It is unclear whether transformation
Geographic Distribution: Worldwide, but different Babesia from merozoite to gametocyte begins in the vertebrate host or
species vary in their geographic distribution in the tick. In the tick midgut, the sexual phase of reproduction
Mode of Transmission: A variety of tick vector species; fight- occurs when the gametocytes fuse to form a zygote. The zygote
ing or biting interactions; vertical transmission; blood invades epithelial cells of the tick gut. The resultant forms, ookine-
transfusion tes, leave the epithelial cell and invade either the salivary gland or
the ovary, where they participate in transstadial and transovarial
Major Clinical Signs: Lethargy, pallor, splenomegaly. Dogs
transmission, respectively. Prior to transmission, another asexual
with severe babesiosis caused by B. canis rossi may show
form of reproduction occurs in the salivary gland, sporogony.
neurologic signs, anuria, tachypnea, and poor perfusion
parameters. Babesiosis in Dogs
Differential Diagnoses: Hemoplasmosis, primary immune- Canine babesiosis is a disease of worldwide importance. Ini-
mediated hemolytic anemia and/or thrombocytopenia, tially, two species of Babesia were recognized in dogs (Babesia
other causes of hemolysis such as oxidative toxins or canis and B. gibsoni); however, now at least nine genetically
microangiopathic hemolysis (e.g., with splenic hemangio- distinct canine piroplasms have been described. Babesia species
sarcoma). Canine monocytic ehrlichiosis, Rocky Mountain that infect dogs vary in their geographic distribution, which in
Spotted Fever, and bacterial endocarditis should also be turn has been subject to change due to movement of infected
considered. animals, movement of tick vectors, and reclassification with
Human Health Significance: Babesia species that infect dogs improved diagnostic techniques. As new species have been
and cats are not known to infect humans. recognized, it has become more important for the clinician to
understand the differences in diagnosis, prognosis, and treat-
ment for infection with each species.

Etiology and Epidemiology Babesia canis


Babesia canis is the most common large Babesia species and
Babesiosis, caused by infection with organisms from the genus has three distinct subspecies: B. canis vogeli, B. canis canis, and
Babesia, is characterized by hemolytic anemia, fever, and sple- B. canis rossi. It has been proposed that these are in fact three
nomegaly. Babesia infections can also be subclinical or cause distinct species (B. vogeli, B. canis, and B. rossi).6 B. canis vogeli
severe life-threatening illness. Babesia spp. are intraerythrocytic is transmitted by the brown dog tick (Rhipicephalus sanguin-
protozoan parasites of the phylum Apicomplexa. Altogether, eus). Infection is most commonly diagnosed in warm, humid
more than 100 species of Babesia have been described, and regions of the world, and the disease occurs throughout the year
with the advent of molecular techniques such as PCR assays, in endemic regions. B. canis vogeli can be found in Africa, Asia,
new species and genotypes are identified each year. Histori- Australia, Europe, and the Americas. In the United States, dis-
cally, species have been named and identified based on the ver- ease is most often diagnosed in the southern regions. B. canis
tebrate host and the size of the parasite (large or small Babesia vogeli reported seroprevalence in dogs has ranged from 3.8% to
species). Large Babesia spp. are 3 to 7 µm in length, whereas 59%.7 The prevalence of seroreactivity is higher in adult dogs
small ­Babesia spp. are 1 to 3 µm in length. In most parts of than in dogs younger than 1 year.8 In an antibody serosurvey
the world, tick vectors are the most important means of trans- of dogs in Florida, 46% of 393 greyhounds were seroreactive.7
mission of Babesia species. However, for some Babesia species, The prevalence of seroreactive dogs within these kennels ranged
such as Babesia gibsoni in North America and Europe, direct from 17% to 100%; the lower prevalence was noted in kennels

727
728 SECTION 4  Protozoal Diseases

Tick ovary Transform into


gametes which Merozoites
Zygote fuse to form released
Ookinete zygotes
migration

Tick
gut

Transstadial
and
transovarial
transmission
Merogony
Rhipicephalus
sanguineus
(binary fission)
Ingested
by tick
Tick salivary vector
gland
Ring form

Sporozoites Sporozoites
Tick penetrate
Sporogony released into feeds
tick saliva dog RBC
on dog
FIGURE 75-1  Life cycle of Babesia canis.

with more intensive tick control. None of 50 non-greyhound, sp. was reported in a dog from Great Britain that never traveled
individually housed adult pet dogs within the same geographic outside of that country.21 The dog died as a result of infection.
area were seroreactive, which implicated environment and breed
susceptibility as possible risk factors in endemic areas.7 Trans- Babesia gibsoni
placental transmission of B. canis vogeli infections is strongly Infections with B. gibsoni occur throughout the world, and the
suspected but unproven in an experimental setting.9 insidious nature of this infection has allowed the inadvertent
B. canis canis is found in Europe and Africa. It is trans- transport of infected dogs from Asia to other areas. B. gibsoni
mitted by the ornate cow tick, Dermacentor reticulatus, but can be found in Africa, Asia, Australia, North and South Amer-
R. sanguineus may also be a vector.10 The incidence of infec- ica, and Europe. Depending on the availability of suitable vec-
tion is highest in the fall and spring. Higher prevalence rates tors, two distinct epidemiologic scenarios exist for B. gibsoni:
are most commonly found in rural or suburban areas that are tick transmission and direct transmission between dogs.22,23
adjacent to prairies or woodlands that provide suitable habi- B. gibsoni is transmitted primarily by the tick vectors Hae-
tat for D. reticulatus.11 B. canis canis infection is most often maphysalis bispinosa and Haemaphysalis longicornis, and
diagnosed in France. Infections are being reported more fre- possibly by R. sanguineus.24-28 In its original endemic area of
quently from other European countries such as Croatia, Poland, Asia, the geographic range of B. gibsoni correlates with that of
and Germany, possibly due to changes in the distribution of H. bispinosa, and tick infestation is a risk factor for infec-
D. ­reticulatus.12 Outbreaks of disease due to B. canis canis infec- tion in nonfighting breeds. In the United States where compe-
tion in Polish sled dogs share many similarities with those due tent tick vectors are not endemic, B. gibsoni infections occur
to B. canis vogeli infection in North American greyhounds.13 in dogs engaged in fighting activities.23,29 Most dogs with
B. canis rossi is transmitted by the yellow dog tick, Haema- B. gibsoni infection in the United States have been American pit
physalis elliptica (formerly Haemaphysalis leachi).14 Reports of bull terriers.29,30 However, an increased prevalence of infection
B. canis rossi infection have been limited to Africa, with the vast has even been noted in fighting breeds in areas such as Japan, where
majority of reports coming from South Africa, where more than Haemaphysalis spp. are endemic. In fighting breeds, transmis-
10% of the dogs evaluated in some veterinary hospitals may sion has been associated with a fight or bite by an infected dog
be affected.15,16 The incidence of infection is highest during the or having been born to an infected bitch. Perinatal transmission
summer months. Breed predispositions for infection have not is believed to occur.3-5
been well studied, but the traditional fighting breeds (­American
pit bull terriers, Staffordshire bull terriers, and bull terriers) Babesia conradae
are more likely to die when they are diagnosed with severe B. conradae (previously referred to as “the California isolate”
babesiosis.17 and the “western piroplasm” and historically misidentified as
B. gibsoni)31 has been detected primarily in dogs from southern
Unnamed Large Babesia Species California.31,32 Tick vectors have not been identified. Trans-
A large Babesia sp. has been isolated from dogs in North mission studies that attempted to prove vector competence for
­America that had been splenectomized or were undergoing R. sanguineus and Dermacentor variabilis were unsuccessful or
chemotherapy for cancer. Infected dogs have been identified in inconclusive.33 The author has identified R. sanguineus as well
North Carolina, New Jersey, New York, and Texas.18-20 A tick as soft-bodied ticks, Ornithodoros coriaceus, on B. conradae–
vector has not been identified. A different novel large Babesia infected dogs. A high prevalence of B. conradae infection was
CHAPTER 75  Babesiosis 729

noted in a kennel of greyhound mixed-breed dogs that were


used to hunt coyotes.34 A history of coyote fights was a risk fac- Box 75-1
tor for infection. B. conradae DNA was detected in the spleen
of 1 of 50 coyotes, so fighting may be one mode of transmission
Clinical Findings in Dogs with Babesiosis
for this Babesia species.
Prevalence Duration
Babesia microti–Like Organism Common signs Hyperacute signs
A B. microti–like parasite (also referred to as Babesia annae Anorexia (primarily B. canis rossi )
or Theileria annae) has been identified in dogs, the majority of Lethargy Hypothermia
which have lived in, or traveled to, northwestern Spain.35-38 Weakness Shock
A single American pit bull terrier confiscated from a suspected Pyrexia Coma
dog-fighting operation in North America was also infected.39 Weight loss Disseminated intravascu-
A high percentage of European and North American foxes are Uncommon/atypical signs lar coagulation
infected with a genetically identical parasite.40,41 A tick vector (predominantly B. canis rossi) Metabolic acidosis
has not been definitively identified, but in Spain an association Ascites Death
between Ixodes hexagonus infestation and B. microti–like infec- Edema Acute signs
tion has been observed.42 Constipation Hemolytic anemia
Diarrhea Icterus
Feline Babesiosis Ulcerative stomatitis Splenomegaly
Feline babesiosis has not been studied as extensively as canine Hemorrhage Lymphadenopathy
babesiosis. Several small Babesia species have been identified Congested mucous Vomiting
in cats, which include Babesia felis, Babesia cati, and Babesia membranes Chronic signs
leo. Babesia felis is a highly pathogenic Babesia species that Polycythemia Intermittent pyrexia
infects domestic cats in southern Africa and the Sudan.43 Infec- Ocular and nasal discharge Partial anorexia
tion of domestic cats has primarily been identified along the Respiratory distress Loss of body condition
coast of South Africa.44,45 Affected cats are usually younger Masticatory myositis Lymphadenomegaly
than 3 years, and there is no recognized breed or sex predilec- Temporomandibular joint pain Splenomegaly
tion. Babesia cati is less pathogenic and found primarily in Back pain No signs
India. Babesia leo has been detected in lions (Panthera leo) Central nervous system signs
in Kruger National Park, as well as a single domestic cat in (seizures, ataxia, paresis)
a molecular survey.46 A small piroplasm was visualized in
blood smears from feral cats in Rio de Janeiro, Brazil, but no
molecular data were available to confirm the identity of these
organisms.47 with other tick- or bloodborne pathogens may also influence the
Large Babesia species also appear to infect cats. DNA clinical signs of disease.
sequences, most similar to those of B. canis canis and a Parasite antigens are incorporated on the erythrocyte surface
B. microti–like parasite, have been amplified from the blood of and induce host-opsonizing antibodies, which in turn leads to
three cats with retroviral infections from Spain and Portugal, removal of infected erythrocytes by the mononuclear-phagocyte
but no organisms were visualized.48 B. canis presentii was iden- system. Soluble parasite antigens can also adhere to the sur-
tified in two cats in Israel.49 Another B. canis–like parasite has face of noninfected erythrocytes and platelets. This may lead
been identified in a naturally infected cat from Poland.50 Feline to their opsonization by antibodies, with or without comple-
babesiosis has not been reported in the United States. ment, and account for hemolytic anemia and thrombocytope-
nia that is often not correlated with the level of parasitemia.
Clinical Features Parasitemia and anemia are more severe in splenectomized dogs,
and splenectomy may precipitate disease in dogs with chronic
Clinical Signs and Their Pathogenesis subclinical infections. Mechanisms other than immune-medi-
Canine Babesiosis ated destruction that contribute to erythrocyte damage include
The pathogenicity of Babesia organisms is determined primarily increased erythrocyte osmotic fragility, direct injury of erythro-
by the species and strain involved. Host factors, such as the age cytes by Babesia parasites, accumulation of cyclic nucleotides,
of the host and the immunologic response generated against the and oxidative injury.52-56 Lipid peroxidation increases eryth-
parasite or vector tick, are also important. Although most often rocyte rigidity and slows the passage of erythrocytes through
subclinically infected, dogs infected with “avirulent” species capillary beds. Soluble parasite proteases activate the kallikrein
such as B. canis vogeli may have severe clinical disease. Simi- system and induce fibrinogen-like protein formation. These pro-
larly, dogs infected with “virulent” species such as B. canis rossi teins make erythrocytes more “sticky,” and they sludge in capil-
may be subclinically infected without overt clinical or labora- lary beds, which contributes to anemia and many of the other
tory findings. However, features that should raise clinical suspi- potential clinical signs. The most severe sludging occurs in the
cion for babesiosis include fever, thrombocytopenia, hemolytic central nervous system (CNS) and muscles.
anemia, and splenomegaly. Dogs often have nonspecific signs Thrombocytopenia may result from immune-mediated
such as lethargy, anorexia, and weakness (Box 75-1). Occa- or coagulatory consumption of platelets. Despite severely
sionally owners note jaundice, mucosal pallor, or discoloration decreased platelet counts, bleeding is rarely observed in dogs
of the urine caused by bilirubinuria or hemoglobinuria. Dogs infected with most Babesia strains, and other abnormal coag-
infected with B. canis vogeli may have fever of unknown origin ulation test results are uncommon (with the exceptions of
only without overt hematologic abnormalities.51 Co-infections B. conradae and also B. canis rossi). Dogs with B. conradae
730 SECTION 4  Protozoal Diseases

infection have experienced life-threatening hemorrhages, the


pathogenesis of which requires further study.57
Other possible complications include membranoprolifera-
tive glomerulonephritis, which may have an immune-mediated
pathogenesis.58-60 Renal failure occurs in up to 40% of dogs
infected with B. microti–like parasites, and their presence has
been associated with increased mortality.35,36,38,61,62 Nonregen-
erative anemia, azotemia, and proteinuria with high urine pro-
tein/creatinine ratios were found in these dogs.35

Severe Babesiosis
Virulent B. canis canis and especially B. canis rossi strains
induce a profound systemic inflammatory response, which can
result in a severe sepsis-like syndrome with multiple organ dys-
function (see Chapter 86). The majority of dogs infected with
B. canis rossi have uncomplicated babesiosis and can be treated
as outpatients. However, up to 31% of the dogs examined at
a university clinic required hospitalization, and 10% of hos-
pitalized dogs did not survive.17 Severe clinical illness in dogs
infected with B. canis rossi results from hypotension, acute
renal failure (ARF), neurologic complications, disseminated
intravascular coagulation (DIC), a hepatopathy, and acute FIGURE 75-2  Greyhound infected with Babesia conradae. Mucosal hemorrhages are
respiratory distress syndrome (ARDS). Tissue hypoxia results apparent. These resolved after treatment with atovaquone and azithromycin. (Courtesy 
Dr. Jane Sykes, University of California, Davis.)
from anemia, shock, vascular stasis, excessive endogenous pro-
duction of carbon monoxide, parasitic damage to hemoglobin,
and decreased ability of hemoglobin to off-load oxygen.53,63 bounding pulses. Fever is not consistently identified, because
Lactic acid generation from tissue hypoxia can result in severe it often waxes and wanes. Tachycardia and tachypnea may be
metabolic acidosis.64 present in severely anemic dogs. Mucosal hemorrhages and/
“Red biliary syndrome” is a paradoxical phenomenon of or epistaxis may be present in dogs with B. conradae infection
severe intravascular hemolysis (manifested as hemoglobinemia (Figure 75-2), and excessive bleeding from venipuncture sites
and hemoglobinuria) in combination with hemoconcentration may be noted.
(high-reference-range or elevated hematocrit).63 The hemocon- Dogs with severe babesiosis may show CNS signs such as
centration is thought to occur when plasma shifts from the vas- incoordination, pelvic limb paresis, muscle tremors, nystag-
cular to the extravascular compartment as a result of increased mus, anisocoria, intermittent loss of consciousness, seizures,
capillary permeability, with a resultant decrease in blood vol- stupor, coma, aggression, or vocalization.63 Dogs with red
ume. Hemoconcentration has been associated with cerebral biliary syndrome may have congested mucous membranes or
babesiosis, DIC, ARF, and ARDS. icterus. Other clinical abnormalities in dogs with severe babe-
Neurologic complications result from sludging of parasitized siosis include tachypnea, increased lung sounds, and cardiac
erythrocytes in CNS capillary beds, with congestion and macro- arrhythmias.
scopic and microscopic hemorrhages. Severe hypoglycemia can Affected cats may be lethargic, pale, tachycardic, and tachy-
also result in neurologic signs. Other complications of severe pneic, and in some cases, icteric.
babesiosis include pancreatitis, rhabdomyolysis, ocular involve-
ment, upper respiratory signs, cardiac arrhythmias, necrosis of Diagnosis
the extremities, and fluid accumulation. Pulmonary, CNS, and
renal complications are associated with a higher rate of mortal- Laboratory Abnormalities
ity. Persistent lactate concentrations above 40 mg/dL are a poor Complete Blood Count
prognostic indicator for survival.65 In dogs with babesiosis, the primary hematologic abnormali-
ties are anemia and thrombocytopenia. Thrombocytopenia is
Feline Babesiosis often present, even when anemia is absent. A mild, normo-
Cats with babesiosis from southern Africa generally show leth- cytic, normochromic anemia is generally noted in the first few
argy, anorexia, weakness, an unkempt haircoat, and/or diar- days after infection, which becomes macrocytic, hypochromic,
rhea.45 Fever and icterus are less common. Anemia can be severe and regenerative as the disease progresses. Uncommonly with
and is the underlying reason for the clinical signs. The disease is B. canis rossi infections, a relative polycythemia may be
chronic, and signs may not be apparent until a later stage of ill- noted.63 Leukocyte abnormalities are inconsistently observed
ness. Cats usually adapt to the anemia and may have only mild but may include leukocytosis (with or without a left shift),
clinical signs until they are stressed by handling. Complications leukopenia, neutrophilia, neutropenia, lymphocytosis, and/
of the hemolytic anemia have included hepatopathy, pulmonary or eosinophilia.34,66-68 Leukopenia or a low-normal leukocyte
edema, renal failure, CNS signs, and concurrent infections. count due to relative neutropenia has been frequently observed
in dogs from Europe with B. canis infections and dogs from the
Physical Examination Findings United States with B. conradae infections.34,68 Autoagglutina-
Physical examination abnormalities in most dogs with babesio- tion, positive direct antiglobulin (Coombs’) tests, and spherocy-
sis consist of fever, mucosal pallor, lethargy, splenomegaly, and tosis may also be present.
CHAPTER 75  Babesiosis 731

TABLE 75-1
Diagnostic Assays Available for Babesiosis in Dogs and Cats

Assay Specimen Type Target Performance


Cell culture Whole blood Babesia spp. Not widely offered or utilized for routine diagnostic
purposes. Requires several weeks’ incubation.
Cytology Whole blood, Babesia spp. Rapid and specific (i.e., when merozoites are identi-
buffy-coat smears fied by experienced cytologists, the sample is likely
(the area just below to be infected with Babesia spp.; however, Babesia
the buffy coat), spp. cannot be accurately differentiated based on
tissue aspirates morphology alone). Less sensitive than PCR.
Immunofluorescent Serum Antibodies to Acute and convalescent serology may be required
antibody serology Babesia spp. for diagnosis of acute infection, because initial
results may be negative in dogs with acute disease
and positive results may reflect previous exposure
rather than active infection. Cross-reactivity can
occur between Babesia spp. Some dogs do not
develop detectable antibody titers despite chronic
infection.
PCR Whole blood, splenic Babesia spp. DNA Confirms active infection. Sensitivity and specificity
aspirates varies depending on assay design and specimen
type. Both false-positive and false-negative results
are possible; PCR results must be interpreted in
light of the clinical signs. Serial sampling (i.e., two
or more tests on specimens obtained 2-4 weeks
apart) will increase sensitivity, especially in chroni-
cally infected animals.

In cats, anemia associated with B. felis infection is typically cases, granular casts. Some dogs have pigmenturia consisting
macrocytic, hypochromic, and regenerative.69 Thrombocytope- most commonly of bilirubin, and occasionally hemoglobin.
nia is an inconsistent finding.
Coagulation Testing
Serum Chemistry Profile The most consistent hemostatic abnormality in complicated and
There are no pathognomonic biochemical findings in dogs with uncomplicated babesiosis is thrombocytopenia. DIC has been
babesiosis. Common findings in North American dogs include reported; however, complete confirmation of DIC in animals
hyperglobulinemia (which may be present in the absence of with babesiosis may be difficult because of the nature of the
other laboratory abnormalities), mildly increased liver enzyme underlying disease process and the reported unreliability of the
activities, and, less commonly, hyperbilirubinemia. A study of human fibrin degradation product test for evaluating canine
dual infections with B. canis and Ehrlichia canis showed that specimens. Dogs with B. conradae infection appear to have
the prevalence of hyperglobulinemia was higher in dogs with altered platelet function tests.57
dual infections than in dogs with a single infection caused by
either organism.70 Hyperbilirubinemia is a consistent finding Microbiologic Testing
during acute disease caused by B. canis canis and B. canis rossi There are three basic methods available for specific diagnosis of
but not by B. gibsoni or B. conradae.34,67 Dogs with severe B. Babesia infections: microscopic identification, serologic testing,
canis rossi infections may have hemoglobinemia, moderately and nucleic acid–based detection methods (Table 75-1). The true
increased liver enzyme activities, increased BUN and serum cre- clinical sensitivity and specificity of most of the available tests is
atinine concentrations, hypoalbuminemia, hypoglycemia, and unknown, but all modalities can have either false-­positive or false-
metabolic acidosis. negative results. Therefore, there is no “perfect” test for Babesia
Cats infected with B. felis typically have elevated serum ALT infection, and in some cases, multiple diagnostic tests are indicated.
activity and total bilirubin concentrations. Serum protein values
are usually within reference limits, but hyperglobulinemia can Cytologic Diagnosis
occur. Renal parameters are unaffected.69 Light microscopic examination is highly specific for the identifi-
cation of Babesia organisms, but because of its limit of detection
Urinalysis (0.001% parasitemia), it has relatively poor sensitivity and is
Urinalysis abnormalities in dogs with babesiosis are variable but not a suitable screening test. Several species/genotypes are vir-
include bilirubinuria, hemoglobinuria, proteinuria, and, in rare tually indistinguishable by light microscopy, making accurate
732 SECTION 4  Protozoal Diseases

identification to the species level impossible. However, when Serologic Diagnosis


cytologic detection is used in combination with the history, sig- Because Babesia parasites are difficult to detect, especially in
nalment, physical examination, clinicopathologic data, and geo- chronic carriers, immunodiagnostics may be used to screen
graphic location, the clinician may be able to predict the most for exposure. Immunofluorescent antibody (IFA) assays are
likely species present. used most commonly to detect antibodies to Babesia species.
Babesia canis are large, pyriform organisms and usually exist Laboratory methods differ, and each laboratory should be con-
singly or in pairs (Figure 75-3, A). Smaller single intracellular sulted for cutoff antibody titers. As a general guideline, titers to
organisms are likely to be B. gibsoni or B. conradae (see Figure B. canis or B. gibsoni of 1:64 or greater on a single specimen are
75-3, B and C). However, when the parasites are rapidly rep- supportive of exposure. Serologic assays for B. conradae are not
licating, the classic intraerythrocytic forms may not predomi- widely available at the time of writing.
nate and some more atypical irregular, amoeboid parasite forms Titers to multiple Babesia species must be measured if anti-
with a wide range of sizes can be detected. During these phases body testing is performed in geographic areas where more than
of infection, some small Babesia spp. have large forms and some one species of Babesia exists. Cross-reactivity between ­Babesia
large Babesia spp. have small forms. In chronic infection, the spp. makes parasite identification by PCR necessary to defini-
level of parasitemia is often low, especially in B. canis–infected tively identify a given species. There are also many documented
dogs, and thorough examination of thin blood smears is nec- instances in which parasites or parasite DNA have been detected
essary. Evaluation of stained slides requires experience and a but anti-Babesia antibodies could not be detected. Very young
significant time commitment on the part of the laboratory tech- dogs or dogs tested early in the disease course may have nega-
nician. For B. canis infections, blood collected from the periph- tive antibody test results, so convalescent serology is required
eral capillary beds of the ear tip or nailbed may yield higher in some cases. Antibodies were not detected in 36% of dogs
numbers of parasitized cells.71 Rarely, phagocytized organisms with B. canis parasitemia in one study.8 ELISAs that target sev-
and erythrocyte fragments are seen in neutrophils. Methods that eral different antigens have been developed for use in antibody
concentrate and stain buffy-coat specimens may improve the or antigen detection tests and offer promise for serodiagno-
sensitivity of parasite detection.72,73 sis of canine babesiosis in the future. However, there are no

A B

C
FIGURE 75-3  Blood smears from dog with babesiosis. A, Large unnamed Babesia piroplasm originally identified in dogs in North Carolina. A pair of large, piriform-shape merozoites
are present within an erythrocyte. B, Individual merozoites of Babesia gibsoni in erythrocytes (arrows). C, Babesia conradae merozoite within an erythrocyte (arrow). Polychromasia and
anisocytosis are also present. Although considered a small Babesia species, in this image, the organism is relatively large. Wright’s stain, 1000×.
CHAPTER 75  Babesiosis 733

commercially available ELISA assays for the detection of anti- vascular injury and poor tissue oxygenation in severely affected
Babesia antibodies in the United States. dogs, are often most severe in the lungs. Nonparasitized cells
often line the endothelial surface with parasitized cells sludged
Molecular Diagnosis Using the Polymerase Chain Reaction in the lumen. Pathologic changes in the brain of these dogs
Genetic methods such as PCR assays are the most sensitive and include congestion, macroscopic and microscopic hemorrhages,
specific means of detecting active infection with Babesia. Some sequestration of parasitized erythrocytes in capillary beds, and
assays have a lower limit of detection that is more than 1300-fold pavementing of parasitized cells against the endothelium. Car-
more sensitive than the level (0.001% parasitized erythrocytes) diac histologic changes include hemorrhage, necrosis, inflam-
of light microscopy.74 Many different PCR assays have been matory infiltrate, and fibrosis. Microthrombi of many tissues
published for the identification and differentiation of Babesia may be evident in animals exhibiting signs of DIC. Impression
infections, and several commercial diagnostic laboratories offer smears of the spleen may substantiate the diagnosis of babesiosis
real-time PCR assays for detection of Babesia infection. Species at necropsy. Nonspecific findings in dogs with all forms of babe-
identification can then be accomplished by DNA sequencing siosis include erythroid hyperplasia in the bone marrow, extra-
or by the use of species-specific PCR assays. PCR assays that medullary hematopoiesis of the liver and spleen, mononuclear
are designed to detect multiple Babesia spp. (i.e., a broad-range phagocyte system hyperplasia, and centrilobular necrosis of the
Babesia PCR) can facilitate screening. A well-designed species- liver. Vasculitis has been observed in B. ­conradae infections and
specific assay will only detect one species, and a negative test is associated with hepatitis and lymphadenitis with multifocal
may lead clinicians to falsely conclude that the patient is not deposits of IgM in inflamed arteries and renal glomeruli.60 In
infected with Babesia because the wrong species was targeted chronic canine babesiosis and feline babesiosis, the only gross
for testing. In addition, broad-range PCR assays have been help- finding may be splenomegaly.
ful for identification of novel Babesia species.
Treatment and Prognosis
Pathologic Findings
The most striking pathologic findings occur in dogs that die Antiprotozoal Treatment
from severe babesiosis (usually associated with B. canis rossi). Dogs
These include staining of tissues with hemoglobin or bilirubin, Dogs generally show clinical improvement within 24 to 72
hepatosplenomegaly, lymphadenomegaly, and kidneys that are a hours of treatment with antibabesial drugs, but some animals
dark-reddish color. Edema and hemorrhage, which may indicate take as long as 7 days to respond (Table 75-2). Imidocarb

TABLE 75-2
Selected Antibabesial Compounds Used in the Treatment of Dogs and Cats

Organism
Interval Duration Babesia Babesia Babesia Babesia
Generic Name* Dose (mg/kg) Route (hours) (days) canis gibsoni conradae felis
Imidocarb 5-6.6 IM Once Repeat in 14 +++ + + —
dipropionate† 7.5 IM Once NA
Diminazene 3.5-5 IM Once‡ NA +++ ++ ? +
aceturate
Azithromycin§ 10 PO 24 10 +++ +++ +++ ?
and
Atovaquone§ 13.3 PO 8 10
Clindamycin‖ 25 PO 12 90 + + ? ?
and
Doxycycline 5 PO 12 90
and
Metronidazole 15 PO 12 90
Primaquine 0.5 PO 24 1-3 ? — ? +++
phosphate 1 mg per cat# IM 36 6 — — ? +++

+++, Very good; ++, good; +, fair to poor; —, not effective; ?, unknown.
*For specific information on each drug, see Chapter 10.
†For combination therapy, a dose of 3.5 mg/kg diminazene has been followed by a dose of 6.0 mg/kg imidocarb.
‡Not available in the United States, except by compassionate use. For B. canis, this dose is sufficient; for B. gibsoni, repeat dose in 24 hours. Total

dosages of 7 mg/kg or higher are associated with an increased risk of neural and parasympathomimetic toxicity.
§These two drugs should be used in combination. Atovaquone must be given with a fatty meal to facilitate absorption. The suspension causes minimal

gastrointestinal side effects.


‖Anecdotal evidence for activity against B. canis. Not effective in clearing the infection. Not recommended as sole therapy because of the potential to

develop resistance. Further documentation needed to establish efficacy of this combination (see text).
#Note that 1 mg/kg is a lethal dose.
734 SECTION 4  Protozoal Diseases

dipropionate is active against B. canis.75 Imidocarb can elimi- can develop or be selected, this author recommends only use
nate B. canis, eliminates infection in ticks that engorge on of the suspension. Dogs that have been splenectomized before
treated animals for up to 4 weeks after treatment, and prevents treatment do not appear to clear infections with treatment.83
infection for up to 6 weeks after a single injection.76 A single Likewise, immunosuppressive therapy may reduce the ability of
dose of 7.5 mg/kg or a single dose of 6 mg/kg given the day after antibabesial treatment to clear dogs of B. gibsoni infection. The
a dose of diminazene (3.5 mg/kg) also clears infection.77 In areas recommended follow-up after treatment with atovaquone and
where reinfection is likely, some practitioners use a lower dose, azithromycin is a minimum of two PCR tests approximately 60
2 mg/kg SC once, that does completely clear infection. This and 90 days after completing treatment.
approach is an attempt to induce a state of premunition. Pre- An alternative treatment strategy can be used to treat
munition is the immunity of existing infection in which chronic B. gibsoni infections that fail to respond to atovaquone and
subclinically affected carriers may be resistant to reinfection or azithromycin.84 This regimen involves a combination of
at least have reduced morbidity with new infections. In contrast, clindamycin, metronidazole, and doxycycline for a minimum
animals that have been cleared of infection are more susceptible of 3 months (see Table 75-2). In an uncontrolled study, 3 of
to reinfection with recurrent clinical disease. The risk of this 4 experimentally infected dogs had negative PCR assay results
approach is that clinical babesiosis may relapse in some chronic after treatment, whereas 1 dog remained persistently infected
carriers. Therefore, this approach is not recommended for the and experienced clinical relapses during treatment.84 Clearance
treatment of canine babesiosis in many parts of the United of the infection as determined by PCR was not detected until
States, where vector transmission and risk for reinfection are 30 and 72 days after completing a 90-day treatment course in
thought to be low. Imidocarb does not clear B. gibsoni infec- 2 dogs. The PCR test result became negative in the third dog
tions, but reduces morbidity and mortality. Therefore, this is 12 days into treatment. Three of the dogs were initially treated
a reasonable alternative treatment for B. gibsoni if the owner with three doses of diminazene aceturate. In another study, 3
cannot immediately afford more effective treatments. The same naturally infected dogs that had clinical relapse after or during
may be true for B. conradae infections. Pretreatment with atro- atovaquone and azithromycin treatment had clinical recoveries
pine (0.5 mg/kg SC 30 minutes before injection) reduces the after treatment with the clindamycin, metronidazole, and dox-
adverse effects of imidocarb (see Chapter 10). ycycline combination.80 The small number of cases reported
Other related aromatic diamidines include diminazene ace- and the high degree of variability in the PCR results after treat-
turate, phenamidine isethionate, and pentamidine isethionate ment make the efficacy of this combination uncertain. How-
(see Chapter 10). Although these are not available for treatment ever, as with the atovaquone-azithromycin combination, PCR
of dogs in the United States, the first two of these drugs have testing should be performed 60 and 90 days after completing
been used in many other countries and are highly active against therapy.
B. canis. They do not clear B. gibsoni infections, but can reduce Aggressive supportive care and monotherapy with clinda-
morbidity and mortality. mycin (25 mg/kg PO q12h for 7 to 21 days) has been recom-
Atovaquone and azithromycin combination therapy is mended if specific antibabesial drugs are not available. At this
the most effective treatment for B. gibsoni and B. conradae dosage, clinical abnormalities resolve even if organism clearance
infections (see Table 75-2).78,79 Atovaquone and azithromy- does not occur. This author does not recommend this practice
cin are given for 10 days. Atovaquone must be administered for B. gibsoni–infected dogs, because it has the potential to
with a fatty meal to maximize drug absorption. The treat- select for macrolide resistance and interfere with subsequent
ment appeared to sterilize B. gibsoni infections or reduce the atovaquone and azithromycin treatment.
parasitemia below detectable limits in approximately 80% of Other treatments that have been used to treat B. canis infec-
dogs in a randomized double-blind placebo-controlled trial.79 tions are quinuronium sulfate and 1% trypan blue solution.
However, in another study, this combination only eliminated Trypan blue does not clear infections and results in bluish dis-
B. gibsoni infection (or reduced parasitemia below detect- coloration of tissues and plasma.
able limits of PCR assay), in 2 of 7 dogs.80 It also did not
clear experimental infection with two Australian isolates of Cats
B. gibsoni.81 When atovaquone is used alone to treat B. gib- Treatment of feline babesiosis has not been as critically evaluated,
soni infection, recrudescence of infection develops more than but most antibabesial drugs appear ineffective. Primaquine phos-
30 days after treatment.81 Drug resistance has been identified phate, an antimalarial compound, administered orally or as an
in vitro, so atovaquone should always be used in combination intramuscular injection, is currently the drug of choice (see Table
with other drugs. Mutations in the genes that encode the puta- 75-2). However, the effective dose, 0.5 mg/kg, is very close to
tive molecular target of atovaquone have been identified in the lethal dose (1 mg/kg). In experimental studies, rifampin and
B. gibsoni isolated from dogs that failed to clear infection after trimethoprim-sulfadiazine were not as effective as primaquine.85
atovaquone treatment.80,82
In this author’s experience, dogs that fail to clear B. gibsoni Supportive Care
infections after initial treatment with atovaquone and azithro- Other supportive care measures that may be required to treat
mycin will not clear their infection after a follow-up treatment animals with babesiosis are packed red blood cell transfusions
with the same drugs, although clinical disease may resolve. and intravenous crystalloid fluid therapy. The administration
There are also two commercially available formulations of of the plasma component of whole blood is unnecessary in the
atovaquone: a single-drug liquid suspension that is very well majority of dogs with babesiosis and can place the patient at
tolerated, and a combination tablet product (atovaquone and risk of volume overload. If rehydration is required, crystalloid
proguanil hydrochloride). Although the combination product replacement solutions are preferable. Whether glucocorti-
is less expensive, it causes severe vomiting in some patients. coids are indicated to counteract the immune-mediated com-
Because drug absorption is critical and resistance to atovaquone plications of babesiosis is controversial. In one study, 21% of
CHAPTER 75  Babesiosis 735

134 dogs with B. canis rossi infection had hemolytic anemias and animals positive by either or both methods should not be
that did not respond to antibabesial therapy alone.63 This used for blood donation.
seems less common with B. canis vogeli or B. gibsoni infec- Of 16,000 greyhounds adopted through rescue leagues in
tions, because these often respond completely to antibabesial 1995, 20% to 60% were seropositive for B. canis. Much of
treatment alone. In most dogs that require glucocorticoids, this screening was done before the availability of PCR assays,
the glucocorticoid dosage can be tapered and discontinued and serologic testing results likely overestimate the true preva-
within a 2- to 3-week period. Continued glucocorticoid ther- lence of infection. The likelihood that an adopted greyhound
apy may predispose the animals to other infections and has will develop clinical babesiosis or directly transmit infection to
the potential to induce ­babesial relapse.86 other dogs is low. However, the risk to other dogs is great if the
infected animal is placed in a breeding kennel in which dogs are
Immunity and Vaccination housed together and tick control is not adequate, or if the ani-
mal is used as a blood donor. Treatment with imidocarb dipro-
The duration of protective immunity against B. canis infec- pionate may eliminate the B. canis carrier status. This approach
tion is limited. Antibody titers may gradually decline between should be considered in situations in which risk of spread is
3 and 5 months after infection.76 Recovered dogs are protected likely. In other situations, the owner should be made aware of
against homologous infection within 5 to 8 months after infec- the seropositive status so that should clinical signs consistent
tion.76 Cross-protection between strains does not occur, and with babesiosis arise, the attending veterinarian can be alerted
seropositivity does not guarantee protection against heterolo- to the possibility of the disease. Splenectomy should be avoided
gous challenge. if possible in these dogs should it become indicated.
A vaccine produced from cell-culture–derived exoantigens
of B. canis canis is available in Europe.87 An efficacy of 70% Public Health Aspects
to 100% has been reported, with the disease occasionally seen
in the vaccinates generally being mild. Other field studies have Canine and feline Babesia spp. do not appear to pose a zoonotic
been less impressive. Vaccination does not prevent infection risk to immunocompetent humans. Their risks to immunocom-
but appears to block initiation of many of the pathologic promised individuals are unknown, but as with many infectious
processes involved in disease pathogenesis.88,89 Vaccines may diseases, people who are undergoing chemotherapy, are infected
limit parasitemia and reduce development of anemia and sple- with HIV, or have been splenectomized should exercise special
nomegaly. A bivalent vaccine derived from soluble parasite caution when handling blood samples from dogs. Babesiosis
antigens from B. canis canis and B. canis rossi (Nobivac Piro, is a rare tick-borne zoonosis of people in Europe and in the
MSD Animal Health South Africa) reduces clinical signs after United States, and isolated uncharacterized Babesia infections
challenge with both species.90,91 Differences in strain antige- have been reported in Africa and Mexico.93 The majority of
nicity limit the usefulness of the commercial vaccine in other infections are mild or asymptomatic; however, some result in
areas. Because B. canis vaccines do not cross protect against all severe illness and death. Splenectomized people or those older
B. canis subspecies, they are highly unlikely to protect against than 55 years are especially at risk.94,95 No Babesia species has
other Babesia species. been identified that is host specific for people. Sylvan cycles with
wild animal reservoirs occur in nature. People serve as acciden-
Prevention tal hosts for Babesia of animals when they are bitten by infected
ticks. Transmission through blood transfusion has also been
Treatment of babesiosis is expensive and may be ineffective, so described. B. microti is the primary parasite that infects people
prevention is of paramount importance. Preventive measures in the northeast and upper Midwest of the United States. The
alone may be sufficient to control B. canis outbreaks in kennels vector tick is Ixodes scapularis, which can co-transmit anaplas-
in the southeastern United States. The primary means of preven- mosis and Lyme borreliosis. The hemolytic disease with flu-like
tion is tick control (see Prevention section in Chapter 28). Early symptoms is usually mild and self-limiting or easily treated with
tick identification and removal is important because it likely clindamycin and quinine. Both asymptomatic infections and
takes a minimum of 2 to 3 days of feeding for transmission of clinical babesiosis with severe anemia have been reported in
the parasite to occur. Dogs should be tested, treated if neces- people in California and Washington and is caused by the more
sary, and quarantined before being introduced into a colony. recently described parasite Babesia duncani.96-98
The use of amitraz-impregnated collars significantly reduced Babesia divergens causes a severe form of human babesio-
new infections in a B. canis rossi–endemic area. In this study sis in Europe, which usually occurs in people who have had a
none of 20 dogs treated with the collars acquired infections, splenectomy and is often fatal. Babesiosis was also identified in
compared to 27% of 30 control dogs that received no tick pre- splenectomized people from Italy and Austria with a new strain
vention.92 Prevention of aggressive interactions with other dogs (EU1) that was more closely related to Babesia odocoilei.98
(especially fighting breeds such as American pit bull terriers) Identical isolates of Babesia have been found in Ixodes ricinus
may also prevent infection. All prospective canine blood donors ticks from Slovenia, suggesting a more widespread distribution
should be tested for babesiosis with serology and PCR assays, of this organism in Europe.
736 SECTION 4  Protozoal Diseases

CASE EXAMPLE Pretreat-


ment
Posttreat- Reference
ment Range
Bartonella <1:16 ND
Signalment: “Dora,” a 4-year-old female spayed American
­vinsonii IFA
Staffordshire terrier from North Carolina
Bartonella <1:16 ND
History: Dora was evaluated for a 3- to 4-day history of anorexia
­henselae IFA
and lethargy. Before referral, an ELISA that detects heartworm
Rickettsia 1:64 ND
antigen and antibodies to E. canis or B. burgdorferi antigens
­rickettsii IFA
was performed and the results were negative. The dog had
SNAP 4Dx ELISA Negative for ND
mild anemia (HCT 28.4%) and moderate thrombocytopenia
all agents
(59,000 platelets/µL) on an automated hematology analyzer.
Babesia spp. PCR Positive Negative
Amoxicillin–clavulanic acid, doxycycline, and famotidine
(B. gibsoni)
were prescribed. The patient began showing signs of
increased energy and appetite as well as an increase in the
HCT (38%) and platelet count (147,000 platelets/µL) after 4 Diagnosis: Babesia gibsoni infection.
weeks of treatment. However, because the values did not Treatment: Atovaquone and azithromycin.
return to within reference range, the case was referred. Comments: Dogs with babesiosis often have a positive
Physical Examination: clinical response to doxycycline, which leads clinicians to
Body Weight: 24.2 kg. suspect rickettsial infection. Although fever is considered
General: Bright, alert, responsive, hydrated. T = 101.6°C a classic sign of babesiosis, most dogs are afebrile during
(38.7°F), HR = 100 beats/min, RR = 24 breaths/min, mucous initial examination, and waxing and waning fever is
membranes pink, CRT <2 s. Physical examination was often documented with serial rectal temperatures.
otherwise unremarkable. Thrombocytopenia is often a more prominent hematologic
Selected Laboratory Findings: finding than anemia and may be the only finding in some
cases. Babesiosis may be associated with changes in both
Pretreat- Posttreat- Reference albumin and globulin concentrations. Hypoalbuminemia
ment ment Range can be secondary to proteinuria (not documented in this
PCV (%) 41 52 39-58 case). Globulins are often increased, although they may not
Platelet count 80 361 190-468 be above the high end of the reference range. Organisms are
(× 103/µL) frequently not observed on routine examination of stained
TP (g/dL) 6.3 6.5 5.2-7.3 blood smears, and PCR assay is required to document
Albumin (g/dL) 2.7 3.8 3-3.9 infection. Panels that detect antibodies to tick-borne
Globulin (g/dL) 3.5 2.7 1.7-3.8 pathogens typically either do not include Babesia spp.
Albumin/globulin 0.78 1.38 0.9-1.8 at all or only detect antibodies against B. canis. Serologic
ratio cross-reactivity between species can occur, leading to
Visualization of Negative Negative inappropriate diagnosis and treatment recommendations.
Babesia on The positive titer to Rickettsia spp. was interesting, but
microscopic in this geographic location, approximately 20% to 25%
examination of of healthy dogs are seropositive to Rickettsia spp., so its
blood smear significance was unclear. Although acute phase serology
Babesia canis IFA 1:256 ND for B. gibsoni was not done, a convalescent titer to B.
Ehrlichia canis <1:16 ND gibsoni at a follow up examination was 1:64.
IFA

SUGGESTED READINGS 3. Abu MIH, Naito I, et al. Babesia infections in puppies: probably
due to transplacental transmission. J Vet Med B Infect Dis Immu-
Birkenheuer AJ, Correa MT, Levy MG, et al. Geographic distribution of nol Food Hyg Vet Public Health. 1973;609:203-206.
babesiosis among dogs in the United States and association with dog 4. Fukumoto S, Suzuki H, Igarashi I, et  al. Fatal experimental
bites: 150 cases (2000-2003). J Am Vet Med Assoc. 2005;227:942-947. transplacental Babesia gibsoni infections in dogs. Int J Parasitol.
Di Cicco MF, Downey ME, Beeler E, et  al. Re-emergence of Babesia 2005;35:1031-1035.
conradae and effective treatment of infected dogs with atovaquone 5. Itoh N, Itoh S. A case of canine babesiosis possibly developed by
and azithromycin. Vet Parasitol. 2012;187:23-27. transplacental infection. J Jpn Vet Med Assoc. 1990;43:275-276.
Jefferies R, Ryan UM, Jardine J, et al. Babesia gibsoni: detection during 6. Uilenberg G, Franssen FF, Perie NM, et al. Three groups of Babe-
experimental infections and after combined atovaquone and azithro- sia canis distinguished and a proposal for nomenclature. Vet Q.
mycin therapy. Exp Parasitol. 2007;117:115-123. 1989;11:33-40.
7. Taboada J, Harvey JW, Levy MG, et al. Seroprevalence of babesiosis
REFERENCES in Greyhounds in Florida. J Am Vet Med Assoc. 1992;200:47-50.
1. Hutcheon D. Diseases amongst dogs. Malignant jaundice or bilious 8. Bobade PA, Oduye OO, Aghomo HO. Prevalence of antibodies
fever of the dog. Agric J Cape Good Hope. 1893;6:476-477. against Babesia canis in dogs in an endemic area. Rev Elev Med
2. Eaton P. Piroplasma canis in Florida. J Parasitol. 1934;20:312-313. Vet Pays Trop. 1989;42:211-217.
CHAPTER 75  Babesiosis 737

9. Harvey JW, Taboada J, Lewis JC. Babesiosis in a litter of pups. 32. Kjemtrup AM, Wainwright K, Miller M, et al. Babesia conradae,
J Am Vet Med Assoc. 1988;192:1751-1752. sp. nov., a small canine Babesia identified in California. Vet Parasi-
10. Cassini R, Zanutto S, Frangipane di Regalbono A, et  al. Canine tol. 2006;138:103-111.
piroplasmosis in Italy: epidemiological aspects in vertebrate and 33. Yamane I, Gardner SR, Telford T, et  al. Vector competence of
invertebrate hosts. Vet Parasitol. 2009;165:30-35. Rhipicephalus sanguineus and Dermacentor variabilis for Ameri-
11. Bourdoiseau G. Canine babesiosis in France. Vet Parasitol. can isolates of Babesia gibsoni. Exp Appl Acarol. 1993;17:913-919.
2006;138:118-125. 34. Dear JD, Owens SD, Biondo A, et al. Babesia conradae infection in
12. Zygner W, Gorski P, Wedrychowicz H. New localities of Derma- coyote hunting dogs infected with multiple blood-borne pathogens.
centor reticulatus tick (vector of Babesia canis canis) in central and In: American College of Veterinary Internal Medicine Forum, New
eastern Poland. Pol J Vet Sci. 2009;12:549-555. Orleans, LA, 2012.
13. Welc-Faleciak R, Rodo A, Sinski E, et al. Babesia canis and other 35. Camacho AT, Guitian EJ, Pallas E, et  al. Azotemia and mortal-
tick-borne infections in dogs in Central Poland. Vet Parasitol. ity among Babesia microti–like infected dogs. J Vet Intern Med.
2009;166:191-198. 2004;18:141-146.
14. Apanaskevich DA, Horak IG, Camicas JL. Redescription of Hae- 36. Camacho AT, Pallas E, Gestal JJ, et  al. Infection of dogs in
maphysalis (Rhipistoma) elliptica (Koch, 1844), an old taxon north-west Spain with a Babesia microti–like agent. Vet Rec.
of the Haemaphysalis (Rhipistoma) leachi group from East and 2001;149:552-555.
southern Africa, and of Haemaphysalis (Rhipistoma) leachi 37. Garcia AT. Piroplasma infection in dogs in northern Spain. Vet
(Audouin, 1826) (Ixodida, Ixodidae). Onderstepoort J Vet Res. Parasitol. 2006;138:97-102.
2007;74:181-208. 38. Zahler M, Rinder H, Schein E, et  al. Detection of a new patho-
15. Collett MG. Survey of canine babesiosis in South Africa. J S Afr Vet genic Babesia microti–like species in dogs. Vet Parasitol.
Assoc. 2000;71:180-186. 2000;89:241-248.
16. Shakespeare AS. The incidence of canine babesiosis amongst sick 39. Yeagley TJ, Reichard MV, Hempstead JE, et al. Detection of Babe-
dogs presented to the Onderstepoort Veterinary Academic Hospi- sia gibsoni and the canine small Babesia “Spanish isolate” in blood
tal. J S Afr Vet Assoc. 1995;66:247-250. samples obtained from dogs confiscated from dogfighting opera-
17. Reyers F, Leisewitz AL, Lobetti RG, et  al. Canine babesiosis in tions. J Am Vet Med Assoc. 2009;235:535-539.
South Africa: more than one disease. Does this serve as a model for 40. Birkenheuer AJ, Horney B, Bailey M, et  al. Babesia microti–like
falciparum malaria? Ann Trop Med Parasitol. 1998;92:503-511. infections are prevalent in North American foxes. Vet Parasitol.
18. Birkenheuer AJ, Neel J, Ruslander D, et al. Detection and molecu- 2010;172:179-182.
lar characterization of a novel large Babesia species in a dog. Vet 41. Criado-Fornelio A, Martinez-Marcos A, Buling-Sarana A, et  al.
Parasitol. 2004;124:151-160. Molecular studies on Babesia, Theileria and Hepatozoon in
19. Holman PJ, Backlund BB, Wilcox AL, et  al. Detection of a large southern Europe. Part I. Epizootiological aspects. Vet Parasitol.
unnamed Babesia piroplasm originally identified in dogs in North 2003;113:189-201.
Carolina in a dog with no history of travel to that state. J Am Vet 42. Camacho AT, Pallas E, Gestal JJ, et  al. Ixodes hexagonus is the
Med Assoc. 2009;235:851-854. main candidate as vector of Theileria annae in northwest Spain. Vet
20. Sikorski LE, Birkenheuer AJ, Holowaychuk MK, et al. Babesiosis Parasitol. 2003;112:157-163.
caused by a large Babesia species in 7 immunocompromised dogs. 43. Penzhorn BL, Schoeman T, Jacobson LS. Feline babesiosis in South
J Vet Intern Med. 2010;24:127-131. Africa: a review. Ann N Y Acad Sci. 2004;1026:183-186.
21. Holm LP, Kerr MG, Trees AJ, et al. Fatal babesiosis in an untrav- 44. Penzhorn BL, Stylianides E, Coetzee MA, et  al. A focus of feline
elled British dog. Vet Rec. 2006;159:179-180. babesiosis at Kaapschehoop on the Mpumalanga escarpment. J S
22. Konishi K, Sakata Y, Miyazaki N, et al. Epidemiological survey of Afr Vet Assoc. 1999;70:60.
Babesia gibsoni infection in dogs in Japan by enzyme-linked immu- 45. Jacobson LS, Schoeman T, Lobetti RG. A survey of feline babesio-
nosorbent assay using B. gibsoni thrombospondin-related adhesive sis in South Africa. J S Afr Vet Assoc. 2000;71:222-228.
protein antigen. Vet Parasitol. 2008;155:204-208. 46. Bosman AM, Venter EH, Penzhorn BL. Occurrence of Babesia felis
23. Matsuu A, Kawabe A, Koshida Y, et al. Incidence of canine Babe- and Babesia leo in various wild felid species and domestic cats in
sia gibsoni infection and subclinical infection among Tosa dogs in Southern Africa, based on reverse line blot analysis. Vet Parasitol.
Aomori Prefecture. Japan. J Vet Med Sci. 2004;66:893-897. 2007;144:33-38.
24. Swaminath CS, Shortt HE. The arthropod vector of Babesia gibsoni. 47. Mendes-de-Almeida F, Faria MC, Branco AS, et al. Sanitary condi-
Indian J Med Res. 1937;25:499-503. tions of a colony of urban feral cats (Felis catus Linnaeus, 1758) in
25. Higuchi S, Fujimori M, Hoshi F, et  al. Development of Babesia a zoological garden of Rio de Janeiro, Brazil. Rev Inst Med Trop
gibsoni in the salivary glands of the larval tick, Rhipicephalus san- Sao Paulo. 2004;46:269-274.
guineus. J Vet Med Sci. 1995;57:117-119. 48. Criado-Fornelio A, Martinez-Marcos A, Buling-Sarana A, et  al.
26. Higuchi S, Izumitani M, Hoshi H, et al. Development of Babesia Presence of Mycoplasma haemofelis, Mycoplasma haemominutum
gibsoni in the midgut of larval tick, Rhipicephalus sanguineus. and piroplasmids in cats from southern Europe: a molecular study.
J Vet Med Sci. 1999;61:689-691. Vet Microbiol. 2003;93:307-317.
27. Higuchi S, Fujimori M, Hoshi F, et al. Observations on the devel- 49. Baneth G, Kenny MJ, Tasker S, et  al. Infection with a proposed
opment of Babesia gibsoni in the midgut of adult female Rhipi- new subspecies of Babesia canis, Babesia canis subsp. presentii, in
cephalus sanguineus ticks. Jpn J Parasitol. 1994;43:308-311. domestic cats. J Clin Microbiol. 2004;42:99-105.
28. Sen SK. The vector of canine piroplasmosis due to Piroplasma gib- 50. Adaszek L, Ukaszewska J, Winiarczyk S, et al. The first case of feline
soni. Indian J Vet Sci Anim Husb. 1933;3:356-363. babesiosis in Poland. Zycie Weterynaryjne. 2008;83:668-670.
29. Birkenheuer AJ, Correa MT, Levy MG, et al. Geographic distribu- 51. Birkenheuer AJ. Unpublished observations, 2012.
tion of babesiosis among dogs in the United States and associa- 52. Hossain MA, Yamato O, Yamasaki M, et  al. Serum from dogs
tion with dog bites: 150 cases (2000-2003). J Am Vet Med Assoc. infected with Babesia gibsoni inhibits maturation of reticulocytes
2005;227:942-947. and erythrocyte 5′-nucleotidase activity in  vitro. J Vet Med Sci.
30. Wang C, Ahluwalia SK, Li Y, et al. Frequency and therapy moni- 2003;65:1281-1286.
toring of canine Babesia spp. infection by high-resolution melting 53. Lobetti RG, Reyers F. Met-haemoglobinuria in naturally occurring
curve quantitative FRET-PCR. Vet Parasitol. 2010;168:11-18. Babesia canis infection. J S Afr Vet Assoc. 1996;67:88-90.
31. Kjemtrup AM, Conrad PA. A review of the small canine piroplasms 54. Makinde MO, Bobade PA. Osmotic fragility of erythrocytes in
from California: Babesia conradae in the literature. Vet Parasitol. clinically normal dogs and dogs infected with parasites. Res Vet
2006;138:112-117. Sci. 1994;57:343-348.
738 SECTION 4  Protozoal Diseases

55. Murase T, Ueda T, Yamato O, et  al. Oxidative damage and 77. Penzhorn BL, Lewis BD, de Waal DT, et al. Sterilisation of Babesia
enhanced erythrophagocytosis in canine erythrocytes infected with canis infections by imidocarb alone or in combination with dimina-
Babesia gibsoni. J Vet Med Sci. 1996;58:259-261. zene. J S Afr Vet Assoc. 1995;66:157-159.
56. Otsuka Y, Yamasaki M, Yamato O, et  al. Increased generation 78. Di Cicco MF, Downey ME, Beeler E, et al. Re-emergence of Babe-
of superoxide in erythrocytes infected with Babesia gibsoni. J Vet sia conradae and effective treatment of infected dogs with atova-
Med Sci. 2001;63:1077-1081. quone and azithromycin. Vet Parasitol. 2012;187:23-27.
57. Sykes JE. Unpublished observations, 2012. 79. Birkenheuer AJ, Levy MG, Breitschwerdt EB. Efficacy of com-
58. Pomianowski A, Lew S, Kuleta Z, et al. Peritoneal dialysis in a dog bined atovaquone and azithromycin for therapy of chronic Babe-
with acute renal failure caused by the infection with Babesia canis. sia gibsoni (Asian genotype) infections in dogs. J Vet Intern Med.
Pol J Natural Sci. 2008;23:257-267. 2004;18:494-498.
59. Suh M, Chung M. Pathogenicity in experimentally infected dogs 80. Sakuma M, Setoguchi A, Endo Y. Possible emergence of drug-resis-
with Babesia gibsoni. Kor J Vet Res. 2000;40:587-599. tant variants of Babesia gibsoni in clinical cases treated with atova-
60. Wozniak EJ, Barr BC, Thomford JW, et al. Clinical, anatomic, and quone and azithromycin. J Vet Intern Med. 2009;23:493-498.
immunopathologic characterization of Babesia gibsoni infection in 81. Jefferies R, Ryan UM, Jardine J, et al. Babesia gibsoni: detection
the domestic dog (Canis familiaris). J Parasitol. 1997;83:692-699. during experimental infections and after combined atovaquone and
61. Camacho AT, Pallas E, Gestal JJ, et  al. Natural infection by a azithromycin therapy. Exp Parasitol. 2007;117:115-123.
Babesia microti–like piroplasm in a splenectomised dog. Vet Rec. 82. Matsuu A, Miyamoto K, Ikadai H, et al. Short report: cloning of
2002;150:381-382. the Babesia gibsoni cytochrome B gene and isolation of three single
62. Guitian FJ, Camacho AT, Telford SR, 3rd. Case-control study of nucleotide polymorphisms from parasites present after atovaquone
canine infection by a newly recognised Babesia microti–like piro- treatment. Am J Trop Med Hyg. 2006;74:593-597.
plasm. Prev Vet Med. 2003;61:137-145. 83. Stegeman JR, Birkenheuer AJ, Kruger JM, et al. Transfusion-asso-
63. Jacobson LS, Clark IA. The pathophysiology of canine babesiosis: ciated Babesia gibsoni infection in a dog. J Am Vet Med Assoc.
new approaches to an old puzzle. J S Afr Vet Assoc. 1994;65:134-145. 2003;222:959-963:952.
64. Leisewitz AL, Jacobson LS, de Morais HS, et al. The mixed acid- 84. Suzuki K, Wakabayashi H, Takahashi M, et  al. A possible treat-
base disturbances of severe canine babesiosis. J Vet Intern Med. ment strategy and clinical factors to estimate the treatment response
2001;15:445-452. in Babesia gibsoni infection. J Vet Med Sci. 2007;69:563-568.
65. Nel M, Lobetti RG, Keller N, et  al. Prognostic value of blood 85. Penzhorn BL, Lewis BD, Lopez-Rebollar LM, et  al. Screening
lactate, blood glucose, and hematocrit in canine babesiosis. J Vet of five drugs for efficacy against Babesia felis in experimentally
Intern Med. 2004;18:471-476. infected cats. J S Afr Vet Assoc. 2000;71:53-57.
66. Omamegbe JO, Uche UE. Haemogram studies in Nigerian local 86. Masuda T, Baba E, Arakawa A. Relapse of canine babesiosis after
dogs suffering from ancylostomiasis, babesiosis, and trypanoso- prednisolone treatment. Mod Vet Pract. 1983;64:931-932.
miasis. Bull Anim Health Prod Afr. 1985;33:335-338. 87. Moreau Y, Vidor E, Bissuel G, et  al. Vaccination against canine
67. Irwin PJ, Hutchinson GW. Clinical and pathological findings of babesiosis: an overview of field observations. Trans R Soc Trop
Babesia infection in dogs. Aust Vet J. 1991;68:204-209. Med Hyg. 1989;83(suppl):95-96.
68. Zygner W, Gojska O, Rapacka G, et al. Hematological changes dur- 88. Schetters TH, Kleuskens J, Scholtes N, et al. Strain variation limits
ing the course of canine babesiosis caused by large Babesia in domes- protective activity of vaccines based on soluble Babesia canis anti-
tic dogs in Warsaw (Poland). Vet Parasitol. 2007;145:146-151. gens. Parasite Immunol. 1995;17:215-218.
69. Schoeman T, Lobetti RG, Jacobson LS, et al. Feline babesiosis: sig- 89. Schetters TP, Kleuskens JA, Scholtes NC, et al. Vaccination of dogs
nalment, clinical pathology and concurrent infections. J S Afr Vet against Babesia canis infection. Vet Parasitol. 1997;73:35-41.
Assoc. 2001;72:4-11. 90. MSD Animal Health South Africa. 2009. http://www.msd-
70. Matthewman LA, Kelly PJ, Bobade PA, et  al. Infections with animal-health.co.za/whats_new/informative_articles/995.aspx.
Babesia canis and Ehrlichia canis in dogs in Zimbabwe. Vet Rec. Last accessed October 20, 2012.
1993;133:344-346. 91. Schetters TP, Kleuskens J, Carcy B, et al. Vaccination against large
71. Bohm M, Leisewitz AL, Thompson PN, et al. Capillary and venous Babesia species from dogs. Parassitologia. 2007;49(suppl 1):13-17.
Babesia canis rossi parasitaemias and their association with out- 92. Last RD, Hill JM, Matjila PT, et al. A field trial evaluation of the
come of infection and circulatory compromise. Vet Parasitol. prophylactic efficacy of amitraz-impregnated collars against canine
2006;141:18-29. babesiosis (Babesia canis rossi) in South Africa. J S Afr Vet Assoc.
72. Mattia AR, Waldron MA, Sierra LS. Use of the Quantitative Buffy 2007;78:63-65.
Coat system for detection of parasitemia in patients with babesio- 93. Kjemtrup AM, Conrad PA. Human babesiosis: an emerging tick-
sis. J Clin Microbiol. 1993;31:2816-2818. borne disease. Int J Parasitol. 2000;30:1323-1337.
73. Comazzi S, Paltrinieri S, Manfredi MT, et al. Diagnosis of canine 94. Persing DH, Herwaldt BL, Glaser C, et  al. Infection with a
babesiosis by Percoll gradient separation of parasitized erythro- Babesia-like organism in northern California. N Engl J Med.
cytes. J Vet Diagn Invest. 1999;11:102-104. 1995;332:298-303.
74. Birkenheuer AJ, Levy MG, Breitschwerdt EB. Development and 95. Falagas ME, Klempner MS. Babesiosis in patients with AIDS:
evaluation of a seminested PCR for detection and differentiation a chronic infection presenting as fever of unknown origin. Clin Infect
of Babesia gibsoni (Asian genotype) and B. canis DNA in canine Dis. 1996;22:809-812.
blood samples. J Clin Microbiol. 2003;41:4172-4177. 96. Quick RE, Herwaldt BL, Thomford JW, et  al. Babesiosis in
75. Adeyanju BJ, Aliu YO. Chemotherapy of canine ehrlichiosis and Washington State: a new species of Babesia? Ann Intern Med.
babesiosis with imidocarb dipropionate. J Am Anim Hosp Assoc. 1993;119:284-290.
1982;18:827-830. 97. Conrad PA, Kjemtrup AM, Carreno RA, et al. Description of Babe-
76. Vercammen F, De Deken R, Maes L. Duration of protective immu- sia duncani n.sp. (Apicomplexa: Babesiidae) from humans and its
nity in experimental canine babesiosis after homologous and heter- differentiation from other piroplasms. J Parasitol. 2006;36:779-789.
ologous challenge. Vet Parasitol. 1997;68:51-55. 98. Herwaldt BL, de Bruyn G, Pieniazek NJ, et al. Babesia divergens–like
infection, Washington State. Emerg Infect Dis. 2004;10:622-629.
CHAPTER 76

Cytauxzoonosis
Leah A. Cohn

schizogenous phase of infection, whereas piroplasms can be


Overview of Cytauxzoonosis found both during illness and in healthy carrier cats.
First Described: Cytauxzoon felis was first described in domes- There is no breed, sex, or age predisposition for infection,
tic cats from Missouri in 19761 and retroviral infection has not been proven to predispose
Cause: Cytauxzoon felis, a hemoprotozoan parasite (phylum cats to infection or disease.9 Often, infected cats are young
Apicomplexa) adults and previously quite healthy. They are typically out-
door or indoor/outdoor cats from wooded suburban or rural
Affected Hosts: Any felid; bobcats are the major reservoir
areas, where they are more likely to encounter a tick that has
host.
recently fed on an infected bobcat (Lynx rufus), the predomi-
Mode of Transmission: Amblyomma americanum (Lone Star nant reservoir host.9,10 Often, multiple cats in a neighborhood
tick), possibly Dermacentor variabilis (American dog tick) are affected nearly simultaneously, which likely reflects the
Geographic Distribution: South central, southeastern, and presence of infected ticks. Infection occurs most commonly in
mid-Atlantic United States, and South America spring and summer when the tick vector is most active.9 Origi-
nally identified in only the south-central United States, the
Major Clinical Signs: Lethargy, anorexia, dyspnea, fever,
geographic range of C. felis has expanded simultaneously with
icterus, anemia
expansion in the range of the vector Lone Star ticks. Infections
Differential Diagnosis: Hemotropic mycoplasmosis, tularemia, in domestic cats are now recognized throughout the south-­
cholangiohepatitis, sepsis, primary immune-mediated central, southeastern, and mid-Atlantic states, and the patho-
hemo­lytic anemia, oxidative toxicities, toxoplasmosis, gen has been identified in bobcats as far north as Pennsylvania
feline infectious peritonitis and North Dakota (Figure 76-2).10,11
Human Health Significance: Cytauxzoon felis is not known to
infect humans. Clinical Features
Signs and Their Pathogenesis
Owners of cats with cytauxzoonosis usually notice an acute
Etiology and Epidemiology onset of profound lethargy and anorexia in a previously healthy
cat. Less consistently, owners report vocalization, elevation of
Cytauxzoonosis, a life-threatening acute febrile illness of cats, the nictitans, tachypnea and/or dyspnea, or icterus, or that the
is caused by the hemoprotozoan parasite Cytauxzoon felis, an cat feels warm to the touch.12 The vast majority of the con-
apicomplexan parasite within the order Piroplasmida and family siderable pathologic damage that occurs during acute illness is
Theileriidae.2 The parasite infects both domestic and wild Feli- due to the schizogenous phase of parasitemia.8 The degree of
dae, but not other mammals.3 Both Amblyomma americanum schizogony appears to be correlated with severity of disease;
(Lone Star ticks) and Dermacentor variabilis (American dog bobcats (the natural host) undergo a brief and milder schizog-
ticks) harbor the pathogen and are competent transmission vec- enous phase as compared to domestic cats, and correspondingly
tors, although it is likely that Amblyomma ticks are the more illness is less pronounced in bobcats than in domestic cats.13
important natural disease vectors.4-6 Schizont-distended mononuclear cells occlude small veins and
As for other apicomplexan parasites, the parasite life cycle capillaries, most notably in the liver, lung, spleen, and lymph
involves sexual reproduction in the vector tick and asexual nodes.8,14 Obstruction fuels hypoxic tissue damage and release
reproduction in the mammalian host. The parasite is inoculated of inflammatory cytokines, which can lead to systemic inflam-
into the host as a sporozoite when an infected tick vector feeds. matory response syndrome, sepsis, disseminated intravascular
As for other members of the Theileriidae family, the pathogen coagulation (DIC), and multisystem organ failure. The presence
exists within the mammalian host in both a non-erythrocytic of piroplasms may trigger hemolysis in the later stages of the
(schizont) and an erythrocytic (piroplasm) form. Approxi- acute illness; however, hemolysis abates in surviving cats, and
mately 2 weeks after inoculation, schizogony occurs, whereby hemolytic anemia has not been noted in chronic carriers.15-18
mononuclear cells become distended with parasites.7,8 When
these cells rupture, merozoites are released and endocytosed by Physical Examination Findings
RBCs, where they are known as piroplasms.8 The piroplasms Physical examination findings associated with acute cytauxzo-
are ingested by naïve ticks during feeding (Figure 76-1). The onosis are nonspecific.8,12,19 The most consistent single finding
acute illness, known as cytauxzoonosis, occurs during the is fever. Although temperatures greater than 104°F (40°C) are

739
740 SECTION 4  Protozoal Diseases

FIGURE 76-1  Life cycle of Cytauxzoon felis. The parasite is maintained in bobcats (top) which are subclinically infected. The sexual cycle of the organism occurs in Amblyomma ameri-
canum hard ticks, which then transmit sporozoites to domestic cats. Sporozoites invade mononuclear cells, where they undergo schizogony. Schizont-filled monocytes obstruct venules and
capillaries and cause multiorgan failure and death. In cats that survive the schizogony phase, the organism infects erythrocytes and forms piroplasms. The importance of domestic cats as a
reservoir for infection of ticks is unknown. The schizogony stage in the bobcat is relatively mild and brief.

FIGURE 76-2  Geographic distribution of Cytauxzoon felis infections. Shaded areas


show confirmed cases in either domestic cats (blue), or bobcats only (ND and PA). The yel-
low region represents the distribution of Amblyomma americanum ticks.

common, body temperature drops in moribund animals, and


many cats are hypothermic for several hours before death.8,12
Cats are almost always lethargic. Icterus and pallor are com-
FIGURE 76-3  Cat with Cytauxzoon felis infection. There is elevation and hyperemia of
mon findings but may not be present initially. The nictitating the nictitating membranes.
membrane is commonly elevated (Figure 76-3). Both tachycar-
dia and tachypnea, with or without increased respiratory effort,
are also common. Cardiac murmurs are sometimes auscultated, Diagnosis
especially in anemic cats. Splenomegaly, hepatomegaly, and
lymphadenomegaly may be appreciable. Cats may have seizures Laboratory Abnormalities
or become obtunded shortly before death. Because the onset of Complete Blood Count
illness occurs as long as 2 weeks after the cat is bitten by a tick, Abnormalities on the CBC in cats with cytauxzoonosis commonly
ectoparasites are not consistently found on examination. include pancytopenia or bicytopenia and characteristic signet ring
CHAPTER 76  Cytauxzoonosis 741

is common in affected cats, but because platelet clumping occurs


readily in cats, the frequency of true thrombocytopenia may be
overestimated.12,19,20 Although not often assayed, prolongation
of APTT and PT in several affected cats suggests that DIC is a
common complication.22,23

Diagnostic Imaging
Imaging studies do not contribute directly to the diagnosis of
cytauxzoonosis, and results are seldom reported in the literature.
Splenomegaly, hepatomegaly, or both may be identified either
on plain radiographs or on abdominal ultrasound examination.
Although thoracic radiographic findings are seldom documented
in the literature, the author has commonly identified diffuse
interstitial pulmonary patterns on radiographs from cats with
respiratory signs. Less commonly, alveolar lung patterns or pleural
effusion are recognized.

Microbiologic Testing
Diagnostic assays currently available for cytauxzoonosis in cats
are described in Table 76-1.

FIGURE 76-4  Wright-Giemsa–stained peripheral blood smear from a cat with Cytologic Examination of Aspirates or Blood Smears
cytauxzoonosis at 1000× magnification. Multiple RBC contain signet ring–shaped piro- Careful microscopic examination for intracellular parasites is
plasms (short arrow); a single RBC demonstrates a tetrad form (long arrow). (Courtesy Dr. the most commonly used method for diagnosis of infection. A
Marlyn Whitney, University of Missouri Veterinary Medical Diagnostic Laboratory.) well-made thin blood smear stained with Wright’s stain or Diff
Quik should be carefully scanned for either RBC piroplasms
intraerythrocytic inclusions (see Cytologic Examination) (Figure or schizont-laden mononuclear cells. Schizont-distended mono-
76-4). Various combinations of neutropenia, lymphopenia, throm- nuclear cells are sometimes, but not usually, found on careful
bocytopenia, and nonregenerative anemia are observed.12,19,20 examination of the entire feathered edge of the blood smear
Occasionally, neutrophilia or bandemia are observed. Although during acute illness (Figure 76-5). If these cells are identified,
thrombocytopenia is usually identified and may be related to DIC a diagnosis of acute cytauxzoonosis is confirmed. More com-
or vasculitis, platelet clumping is common even in healthy cats monly, microscopic review allows identification of piroplasms
and can result in false thrombocytopenia. Anemia may be absent within RBCs. Unfortunately, because illness is most associated
at the onset of illness but usually develops about the same time with schizogenous replication, illness precedes development of
piroplasms become identifiable. Because of the acute nature of the piroplasms in up to half of cats examined. When cytauxzoono-
disease, anemia is initially nonregenerative. In recovered carrier sis is suspected but piroplasms are not identified, reexamination
cats, cell counts rebound to values within reference ranges, and of another blood smear the following day may yield positive
chronic anemia is not a feature of the carrier state.15,17,18,21 results. Piroplasms are most often 1 to 1.5 µm, signet ring–
shaped inclusions, but tetrad, “safety pin,” and coccoid organ-
Serum Chemistry Profile isms are seen on occasion (see Figures 76-4 and 76-5).8 Stain
Although there are no specific findings on serum chemistry precipitate, Howell-Jolly bodies, or other RBC pathogens such
analysis, a number of abnormalities are commonly identified in as Mycoplasma haemofelis have features that distinguish them
cats with cytauxzoonosis. Hyperbilirubinemia is common and from C. felis piroplasms, but a less experienced microscopist
may result from hepatic vascular occlusion and/or hemolysis. may misidentify them as C. felis. Importantly, recovered cats
The activities of serum ALP and ALT increase in a minority of may harbor piroplasms for months to years after initial infection.
sick cats, and increases are usually mild to moderate.12 Mild to It is important to realize that piroplasms might be discovered
moderate hyperglycemia is common in stressed, sick cats. Mild incidentally in chronic carrier cats that survived infection in the
hypoproteinemia, hypocalcemia, hyponatremia, and hypokale- distant past. Thus, although identification of piroplasms con-
mia can occur during acute illness.12,19 No consistent abnor- firms infection, piroplasms alone (especially in low numbers)
malities have been reported on serum chemistry analysis for cannot confirm that current illness is due to cytauxzoonosis.
recovered carrier cats.17,21 When compared with peripheral blood, mononuclear cells
distended with merozoites are more commonly identified on
Urinalysis fine-needle aspiration (FNA) from spleen, liver, lymph nodes, or
Urinalysis results are rarely reported for cats with cytauxzoono- lungs of affected cats. Because schizogony precedes piroplasm
sis.19 Certainly, hyperbilirubinemia may be reflected by bilirubi- development, FNA of these organs can rapidly confirm a diag-
nuria, and hyperglycemia that surpasses the renal threshold may nosis when piroplasms are absent (Figure 76-6).
cause mild glucosuria. In the author’s experience, the urinalysis
is otherwise unremarkable. Molecular Diagnosis using the Polymerase Chain Reaction
PCR assays are the most sensitive diagnostic test for cytauxzoono-
Clotting Function sis.18,24 Several commercial veterinary diagnostic laboratories that
Acute cytauxzoonosis is a form of sepsis and, as such, is expected perform molecular diagnostics offer PCR analysis of whole blood
to be associated with altered coagulability. Thrombocytopenia samples for Cytauxzoon felis DNA. As with any PCR test, only
742 SECTION 4  Protozoal Diseases

TABLE 76-1
Diagnostic Assays Currently Available for Cytauxzoonosis in Cats
Assay Specimen Type Target Performance
Cytologic examination Whole blood smears, Cytauxzoon felis piroplasms in Piroplasms may initially not be visible in
aspirates of lymph nodes, erythrocytes, or merozoites/ acutely ill cats or may be confused with
spleen, liver, lung schizonts in mononuclear cells other erythrocyte inclusions such as
or reticuloendothelial tissues hemoplasmas or Howell-Jolly bodies.
Schizonts may be found in fine-needle
aspirates of reticuloendothelial tissues
before piroplasms are found on blood
smear examination in some cats.
PCR assays Whole blood C. felis DNA Sensitivity and specificity may vary
depending on assay design, but PCR
assays may be more sensitive than
cytology for detection of piroplasms,
especially in chronic carrier cats.

FIGURE 76-5  Wright-Giemsa–stained peripheral blood smear from a cat with


cytauxzoonosis at 1000× magnification. A mononuclear cell massively distended with a
schizont is surrounded by multiple RBCs, a few of which contain Cytauxzoon felis piroplasms.

FIGURE 76-6  Fine-needle aspirate of a lymph node from a cat with cytauxzoonosis.
reputable laboratories should be used; quality control is crucial. Large numbers of merozoites (which have a granular appearance) erupt from a mono-
Because of the delay inherent in any mail-out PCR test, presump- nuclear cell.
tive disease treatment needs to precede the results of PCR testing.
Healthy, chronic carrier cats have been identified by PCR
assays applied to peripheral blood samples.18,25,26 Although the Pathologic Findings
importance of such carrier cats in propagation of infection is Gross Pathologic Findings
not known, they are competent hosts for transmission of infec- Icterus and pallor are common gross pathologic findings in cats
tion to other cats via the bite of a tick vector.27 Some cat owners that die from cytauxzoonosis.8,19,31 The spleen and liver are
may request screening of healthy cats that reside in the same often enlarged, and mesenteric lymph nodes may be enlarged
neighborhood as a cat with cytauxzoonosis; such screening is and edematous.8,19,31 The lungs are often wet and fail to col-
best accomplished with PCR assays. Unfortunately, no method lapse. Pleural effusion is sometimes identified, as is pericardial
has been confirmed to completely eliminate parasitemia from effusion.32 Petechial hemorrhages are occasionally found on the
chronic carrier cats.21,28,29 serosal surface of organs.8,33
Although histopathology is not a commonly performed ante-
Other Diagnostic Assays mortem diagnostic test, a diagnosis of cytauxzoonosis is easily
To date, C. felis has not been successfully cultured in  vitro. confirmed at necropsy by histopathology.19,31,33 Numerous
Although serologic tests have been developed for detection of venules and capillaries, especially in the spleen, liver, and lungs,
antibodies to C. felis, they are not practical for disease diagnosis, are occluded with markedly enlarged merozoite-distended
because illness likely precedes antibody formation.30 mononuclear cells (Figure 76-7).19,33,34 The germinal centers of
CHAPTER 76  Cytauxzoonosis 743

TABLE 76-2
Antimicrobial Drugs Indicated for Treatment of Feline
Cytauxzoonosis
Dose Interval Duration
Drug (mg/kg) Route (hours) (days)
Atovaquone/azithro-
mycin combination:
Atovaquone and 15 PO 8 10
Azithromycin 10 PO 24 10

Supportive Care
Cats with cytauxzoonosis are critically ill and require support-
FIGURE 76-7  Histopathologic image of a section of spleen from a cat that died of ive care. Crystalloid fluids address dehydration, correct prerenal
cytauxzoonosis. Schizont-distended mononuclear cells obstruct blood vessels. H&E stain, azotemia, and may improve organ perfusion but should be used
400× magnification. (Courtesy Dr. Linda Berent, University of Missouri Veterinary Medical judiciously in cats with evidence of pulmonary damage or pleural
Diagnostic Laboratory.) effusion. Whole blood or packed RBC transfusion is often necessary
during the hemolytic stage of acute illness. Plasma administration
lymphoid tissues contain many parasitized mononuclear cells may be useful in cats with DIC, and although the recommendation
as well, but organisms are not found in lymphocytes them- is controversial, some clinicians advocate treatment with heparin
selves.8 Pulmonary lesions, which are common, are typified at 200 units/kg SC q8h.12 Because affected cats may be severely
by moderate to severe vascular occlusion of medium-caliber febrile and apparently in pain, antipyretic, anti-inflammatory,
vessels and interstitial pneumonia with edema and neutrophil and analgesic drugs are often used. Unfortunately, no controlled
accumulation.35 studies have examined whether these therapies improve or worsen
clinical outcome. The author does not usually administer either
Treatment and Prognosis glucocorticoids or nonsteroidal anti-inflammatory drugs but has
treated cats with buprenorphine as an analgesic. There are numer-
Antiprotozoal Drugs ous reports of antibacterial drugs used in the therapy of cats with
Currently, the treatment of choice for feline cytauxzoonosis cytauxzoonosis, but the drugs utilized (enrofloxacin, doxycycline,
is a combination of atovaquone suspension (Mepron, Glaxo- and/or sodium ampicillin) seem unlikely to have contributed sub-
SmithKline, Research Triangle Park, NC) and azithromycin stantially to any improved outcome.19,37 Cats typically have a poor
(see also Chapter 10) (Table 76-2). Both drugs are adminis- appetite during recovery from cytauxzoonosis. Early placement of
tered orally for 10 days.12 Although atovaquone is expensive, a nasoesophageal or esophagostomy feeding tube not only allows
a single bottle will treat many cats. Compounding pharmacies for provision of enteral nutrition and hydration, but facilitates
may supply reasonably priced atovaquone in smaller quanti- administration of oral medications (i.e., atovaquone and azithro-
ties. In recovered carrier cats, the combination of atovaquone mycin) with minimal stress to the patient.
and azithromycin does not consistently eliminate parasitemia,
but in most treated cats, pathogen burden drops to levels below Prognosis
PCR detection and piroplasms are no longer visible on periph- In the early history of the disease, treatment options for cytaux-
eral blood smears.19 zoonosis were considered ineffective and the disease was
Other antiprotozoal therapies have been investigated for the regarded as uniformly fatal. However, it is now recognized that
treatment of cytauxzoonosis. Parvaquone and buparvaquone, some cats become infected without recognized clinical illness,
both used to treat related Theileria infections, were ineffective and others survive illness with supportive or specific medical
for treatment of experimental cytauxzoonosis.36 Imidocarb care.12,15,18,26,38 Either differences in the individual cat’s immune
dipropionate has not proved to be an effective treatment for response to infection or reduced pathogenicity of the infecting
cytauxzoonosis; only 25% of cats treated with the drug sur- organism are possible explanations for chronic infection in cats
vived.12 Additionally, imidocarb does not reduce the parasite with no history of prior illness. The latter theory is supported
burden in carrier cats.29 Diminazene aceturate is a drug used by finding many cats with chronic infection but no history of
extensively in Africa, South America, and the far East for treat- illness in a limited geographic region around northwestern
ment of protozoal diseases. In a retrospective report, 5 of 6 cats Arkansas.15,27,39
treated with diminazene aceturate at 2 mg/kg IM, plus support- Despite occasional recognition of subclinically infected cats,
ive care, survived acute cytauxzoonosis.22 The author has used most infected cats become extremely ill and die in the absence of
diminazene to treat four additional cats with cytauxzoonosis, aggressive treatment.19,39 Even when treated with atovaquone
and only one survived. Studies in naturally infected cats are and azithromycin, only 60% of infected cats survive.12 Because
difficult to conduct because this drug is not approved for use most cats that die despite therapy do so within a day of pre-
in the United States. As with imidocarb, diminazene aceturate sentation, even the most active antiprotozoal compound may
was unable to eliminate or reduce parasitemia from healthy not prevent death in a proportion of infected cats once clinical
carrier cats.21,30 illness is recognized.
744 SECTION 4  Protozoal Diseases

Immunity and Vaccination selamectin, although the latter does not make a label claim of
efficacy against ticks in cats.44,45 Given their potent activity
Cats that survive experimentally induced schizogenous infection against ticks and their safety in cats, flumethrin-impregnated
appear to be immune to illness on re-infection, although experi- collars offer the greatest potential for prevention of cytauxzo-
mental inoculation with piroplasms alone does not provide onosis in cats that reside in endemic areas. However, no ecto-
protection.36,40,41 Resistance to re-infection with C. felis offers parasiticide prevents tick bites entirely.12,46 Where possible,
hope that successful vaccine development may be possible, but cats in endemic areas should be kept indoors to minimize tick
safe and effective vaccines against apicomplexan parasites are exposure.
notoriously difficult to develop.42 Inoculation of cats with a Recovered cats may remain carriers, and may serve as reser-
less pathogenic Cytauxzoon species (C. manul) failed to reduce voirs for infection of naïve ticks.6 For this reason, recovered cats
mortality associated with C. felis challenge.43 Many veterinar- should also be kept indoors and ectoparasite control practices
ians in endemic regions have observed clinical illness indistin- instituted in order to minimize the risk of transmission to other
guishable from cytauxzoonosis in cats known to have survived cats via tick bite. Treatment of recovered cats with imidocarb
cytauxzoonosis in previous years. Because no attempt has been dipropionate or diminazene aceturate does not reduce piro-
made to look for schizonts in the cats experiencing a second ill- plasm parasitemia, but treatment with atovaquone and azithro-
ness, it is unclear if these cats have recrudescent cytauxzoonosis, mycin may reduce the parasite burden and potentially reduce
re-infection, or some other illness altogether. the risk of transmission to feeding ticks.29,30

Prevention Public Health Aspects


For now, prevention relies on avoidance of tick exposure. Given Cytauxzoon felis is not known to infect humans. However,
the sensitivity of domestic cats to many commonly used ecto- infected cats have the potential to be infected with other vector-
parasiticides, the arsenal of acaricides available for cats is lim- borne pathogens, so precautions should be taken to avoid needle-
ited to flumethrin-impregnated collars, fipronil, and possibly stick injuries or direct skin contact with blood from infected cats.

CASE EXAMPLE maintenance rate. Heparin (200 units/kg SC q8h) and


buprenorphine (0.01 mg/kg SC q8h) were administered.
The following day, the cat’s PCV was 20% and icterus was
Signalment and History: “Ginger,” a 2-year-old female evident on physical examination. Additionally, many
spayed domestic longhaired cat that lived near Springfield, piroplasms were visible on cytologic analysis of a smear
Missouri, was evaluated for a 1-day history of anorexia and of peripheral blood. A serum chemistry profile confirmed
lethargy in April of the year. The cat was an indoor/outdoor hyperbilirubinemia and a mild increase in the activity of
cat with no known history of tick exposure. There were no serum ALT but was otherwise unremarkable.
other cats in the household. By day 3, the PCV was 12%, and both respiratory and heart rate
Physical Examination: On examination, the cat’s rectal were increased. The cat was administered a single unit of
temperature was 105.6°F (40.9°C), with a heart rate of 200 whole blood, after which the PCV increased to 19%. Nutri-
beats/min and a respiratory rate of 28 breaths/min. The tional support was administered via the nasoesophageal
spleen was prominent on abdominal palpation and the tube, but otherwise treatment remained unchanged.
nictitans was elevated (see Figure 76-3), but examination On day 5, the PCV dropped to 11%, which necessitated another
was otherwise unremarkable. whole blood transfusion. The cat remained tachypneic after
Laboratory Findings: Blood was obtained for a CBC and transfusion. Thoracic radiographs revealed pleural effusion
retrovirus testing. The cat tested negative for FeLV antigen and a marked diffuse interstitial lung pattern. The cat was
and FIV antibody. The cat had a mild, nonregenerative treated with supplemental oxygen, and 25 mL of a clear, yel-
anemia (PCV 27%) and leukopenia (4200 cells/µL, reference low, modified transudate was removed using thoracocentesis.
range 5500-19,500 cells/µL). Although the platelet count By day 7, the cat no longer required supplemental oxygen to
was low, platelet clumps were present. Plasma in the spun maintain respiratory function, and the anemia was regen-
hematocrit tube was icteric. No parasites were observed on erative. The cat was discharged from the hospital on day
microscopic slide review. 8 with continued administration of atovaquone and azithro-
Because of a high degree of suspicion for cytauxzoonosis, a mycin at home, but all other treatments were discontinued.
fine-needle aspirate was obtained from the spleen. Multiple On recheck examination 1 week later, the cat was seemingly
markedly enlarged mononuclear cells containing numerous normal to the owners, and no abnormalities were detected
merozoites were observed. on physical examination. The PCV was 29%, platelet and
Diagnosis: Cytauxzoonosis. WBC counts had normalized, and a single piroplasm was ob-
Treatment and Outcome: A nasoesophageal tube served on careful review of the entire feathered edge of the
was placed to facilitate administration of atovaquone blood smear. The cat’s owners were advised to keep the cat
suspension (15 mg/kg PO q8h) and azithromycin (10 entirely indoors and to use an ectoparasiticide regularly to
mg/kg PO q24h). A cephalic vein catheter was placed minimize any risk that the cat could serve as a reservoir for
and intravenous crystalloid fluids administered at a infection of ticks. The cat remained healthy 1 year later.
CHAPTER 76  Cytauxzoonosis 745

SUGGESTED READINGS 19. Hoover JP, Walker DB, Hedges JD. Cytauxzoonosis in cats: eight
cases (1985-1992). J Am Vet Med Assoc. 1994;(205):455-460.
Brown HM, Lockhart JM, Latimer KS, et al. Identification and genetic 20. Franks PT, Harvey JW, Shields RP, et al. Hematological findings
characterization of Cytauxzoon felis in asymptomatic domestic cats in experimental feline cytauxzoonosis. J Am Anim Hosp Assoc.
and bobcats. Vet Parasitol. 2010;(172):311-316. 1988;24:395-401.
Cohn LA, Birkenheuer AJ, Brunker JD, et al. Efficacy of atovaquone and 21. Lewis KM, Cohn LA, Marr H, et  al. Diminazene diaceturate for
azithromycin or imidocarb dipropionate in cats with acute cytauxzo- the treatment of chronic Cytauxzoon felis parasitemia in naturally
onosis. J Vet Intern Med. 2011;25:55-60. infected cats. J Vet Intern Med. 2012;26:1490-1493.
Reichard MV, Baum KA, Cadenhead SC, et  al. Temporal occurrence 22. Greene CE, Latimer K, Hopper E, et al. Administration of dimina-
and environmental risk factors associated with cytauxzoonosis in zene aceturate or imidocarb dipropionate for treatment of cytaux-
domestic cats. Vet Parasit. 2008;152:314-320. zoonosis in cats. J Am Vet Med Assoc. 1999;215:497-500.
23. Garner MM, Lung NP, Citino S, et  al. Fatal cytauxzoono-
REFERENCES sis in a captive-reared white tiger (Panthera tigris). Vet Pathol.
1. Wagner JE. A fatal cytauxzoonosis-like disease in cats. J Am Vet 1996;33:82-86.
Med Assoc. 1976;168(7):585-588. 24. Birkenheuer AJ, Marr H, Alleman AR, et  al. Development and
2. Lack JB, Reichard MV, Van Den Bussche RA. Phylogeny and evo- evaluation of a PCR assay for the detection of Cytauxzoon felis
lution of the piroplasmida as inferred from 18S rRNA sequences. DNA in feline blood samples. Vet Parasitol. 2006;137:144-149.
Int J Parasitol. 2012;42:353-363. 25. Haber MD, Tucker MD, Marr HS, et al. The detection of Cytauxzoon
3. Kier AB, Wightman SR, Wagner JE. Interspecies transmission of felis in apparently healthy free-roaming cats in the USA. Vet Parasitol.
Cytauxzoon felis. Am J Vet Res. 1982;43:102-105. 2007;146:316-320.
4. Blouin EF, Kocan AA, Glenn BL, et al. Transmission of Cytauxzoon 26. Lewis KM, Cohn LA, Downey M, et al. Evaluation of Cytauxzoon
felis Kier, 1979 from bobcats, Felis rufus (Schreber), to domestic felis infection status in captive-born wild felids housed in areas endemic
cats by Dermacentor variabilis (Say). J Wildl Dis. 1984;20:241-242. for this pathogen. J Am Vet Med Assoc. 2012;241:1088-1092.
5. Bondy Jr PJ, Cohn LA, Tyler JW, et al. Polymerase chain reaction 27. Reichard MV, Meinkoth JH, Edwards AC, et al. Transmission of
detection of Cytauxzoon felis from field-collected ticks and sequence Cytauxzoon felis to a domestic cat by Amblyomma americanum.
analysis of the small subunit and internal transcribed spacer 1 Vet Parasitol. 2009;161:110-115.
region of the ribosomal RNA gene. J Parasitol. 2005;91:458-461. 28. Cohn LA, Birkenheuer AJ, Ratcliff E. Comparison of two drug pro-
6. Reichard MV, Edwards AC, Meinkoth JH, et al. Confirmation of tocols for clearance of Cytauxzoon felis infections [Abstract]. J Vet
Amblyomma americanum (Acari: Ixodidae) as a vector for Cytaux- Intern Med. 2008;22:704.
zoon felis (Piroplasmorida: Theileriidae) to domestic cats. J Med 29. Lewis KM, Cohn LA, Marr H, et al. High dose diminazene diacetu-
Entomol. 2010;47:890-896. rate does not eliminate parasitemia in cats with chronic Cytauxzoon
7. Simpson CF, Harvey JW, Lawman MJ, et  al. Ultrastructure of felis infection. Proceedings of the American College of Veterinary
schizonts in the liver of cats with experimentally induced cytauxzo- Internal Medicine Forum. New Orleans: LA; 2012.
onosis. Am J Vet Res. 1985;46:384-390. 30. Cowell RL, Fox JC, Panciera RJ, et al. Detection of anticytauxzoon
8. Kier AB, Wagner JE, Kinden DA. The pathology of experimental antibodies in cats infected with a Cytauxzoon organism from bob-
cytauxzoonosis. J Comp Pathol. 1987;97:415-432. cats. Vet Parasitol. 1988;28:43-52.
9. Reichard MV, Baum KA, Cadenhead SC, et al. Temporal occurrence 31. Aschenbroich SA, Rech RR, Sousa RS, et  al. Pathology in
and environmental risk factors associated with cytauxzoonosis in practice. Cytauxzoon felis infection. J Am Vet Med Assoc.
domestic cats. Vet Parasitol. 2008;152:314-320. 2012;240:159-161.
10. Shock BC, Murphy SM, Patton LL, et al. Distribution and preva- 32. Butt MT, Bowman D, Barr MC, et al. Iatrogenic transmission of
lence of Cytauxzoon felis in bobcats (Lynx rufus), the natural Cytauxzoon felis from a Florida panther (Felix concolor coryi) to a
reservoir, and other wild felids in thirteen states. Vet Parasitol. domestic cat. J Wildl Dis. 1991;27:342-347.
2011;175:325-330. 33. Wagner JE, Ferris DH, Kier AB, et  al. Experimentally induced
11. Birkenheuer AJ, Marr HS, Warren C, et  al. Cytauxzoon felis cytauxzoonosis-like disease in domestic cats. Vet Parasitol.
infections are present in bobcats (Lynx rufus) in a region where 1980;6:305-311.
cytauxzoonosis is not recognized in domestic cats. Vet Parasitol. 34. Susta L, Torres-Velez F, Zhang J, et al. An in situ hybridization and
2008;153:126-130. immunohistochemical study of cytauxzoonosis in domestic cats.
12. Cohn LA, Birkenheuer AJ, Brunker JD, et  al. Efficacy of atova- Vet Pathol. 2009;46:1197-1204.
quone and azithromycin or imidocarb dipropionate in cats with 35. Snider TA, Confer AW, Payton ME. Pulmonary histopathology of
acute cytauxzoonosis. J Vet Intern Med. 2011;25:55-60. Cytauxzoon felis infections in the cat. Vet Pathol. 2010;47:698-702.
13. Blouin EF, Kocan AA, Kocan KM, et al. Evidence of a limited schi- 36. Motzel SL, Wagner JE. Treatment of experimentally induced
zogenous cycle for Cytauxzoon felis in bobcats following exposure cytauxzoonosis in cats with parvaquone and buparvaquone. Vet
to infected ticks. J Wildl Dis. 1987;23:499-501. Parasitol. 1990;35:131-138.
14. Wagner JE. A fatal cytauxzoonosis-like disease in cats. J Am Vet 37. Walker DB, Cowell RL. Survival of a domestic cat with naturally
Med Assoc. 1976;168:585-588. acquired cytauxzoonosis. J Am Vet Med Assoc. 1995;206:1363-1365.
15. Meinkoth J, Kocan AA, Whitworth L, et al. Cats surviving natural 38. Brown HM, Lockhart JM, Latimer KS, et  al. Identification and
infection with Cytauxzoon felis: 18 cases (1997-1998). J Vet Intern genetic characterization of Cytauxzoon felis in asymptomatic
Med. 2000;14:521-525. domestic cats and bobcats. Vet Parasitol. 2010;172:311-316.
16. Kocan AA, Blouin EF, Glenn BL. Hematologic and serum chemical 39. Brown HM, Berghaus RD, Latimer KS, et al. Genetic variability of
values for free-ranging bobcats, Felis rufus (Schreber), with reference Cytauxzoon felis from 88 infected domestic cats in Arkansas and
to animals with natural infections of Cytauxzoon felis Kier, 1979. Georgia. J Vet Diagn Invest. 2009;21:59-63.
J Wildl Dis. 1985(21):190-192. 40. Glenn BL, Kocan AA, Blouin EF. Cytauxzoonosis in bobcats. J Am
17. Lewis KM, Cohn LA, Birkenheuer AJ. Lack of evidence for perina- Vet Med Assoc. 1983;183:1155-1158.
tal transmission of Cytauxzoon felis in domestic cats. Vet Parasitol. 41. Ferris DH. A progress report on the status of a new disease of
2012;188:172-174. American cats: cytauxzoonosis. Comp Immun Microbiol Infect Dis.
18. Brown HM, Latimer KS, Erikson LE, et al. Detection of persistent 1979;1:269-276.
Cytauxzoon felis infection by polymerase chain reaction in three 42. Sharma S, Pathak S. Malaria vaccine: a current perspective. J Vector
asymptomatic domestic cats. J Vet Diagn Invest. 2008;20:485-488. Borne Dis. 2008;45:1-20.
746 SECTION 4  Protozoal Diseases

43. Joyner PH, Reichard MV, Meinkoth JH, et  al. Experimental 45. Stanneck D, Kruedewagen EM, Fourie JJ, et al. Efficacy of an imi-
infection of domestic cats (Felis domesticus) with Cytauxzoon docloprid/flumethrin collar against fleas and ticks on cats. Parasit
manul from Pallas’ cats (Otocolobus manul). Vet Parasitol. Vectors. 2012;5:82.
2007;146:302-306. 46. Dryden MW. Flea and tick control in the 21st century: challenges
44. Bishop BF, Bruce CI, Evans NA, et  al. Selamectin: a novel and opportunities. Vet Dermatol. 2009;20:435-440.
broad-spectrum endectocide for dogs and cats. Vet Parasitol.
2000;91:163-176.
CHAPTER 77

Canine and Feline Hepatozoonosis


Nancy Vincent-Johnson

in 1978 in dogs from the Gulf Coast region of Texas and has
Overview of Canine Hepatozoonosis subsequently been detected in dogs from Louisiana, Oklahoma,
First Described: Hepatozoon canis, India, 1905 (James)1; Hepa- Alabama, Georgia, Florida, and Tennesee.2,21-25 In 1997, it was
tozoon americanum, USA, 1978 (Craig and others)2 identified as a distinct species, H. americanum.26,27 Occasional
Cause: Hepatozoon canis, Hepatozoon americanum (phylum cases have been identified in diverse locations across the United
Apicomplexa) States, including Washington, California, Nebraska, Vermont,
and Virginia.17 Because infection is chronic, the infection may
Affected Hosts: Domestic dogs, possibly some wild carnivores
be diagnosed in dogs with a history of travel to endemic areas.
Geographic Distribution: H. americanum, southeastern and The DNA of a Hepatozoon felis–like organism was detected in
south-central United States; H. canis, Asia, Africa, southern a dog from southern California, but the clinical significance and
Europe, the Middle East, the Americas. epidemiology of this organism are unknown.28
Mode of Transmission: Primarily by ingestion of tick vectors. Recent reports indicate that H. canis also exists in the United
For H. americanum, the vector is Amblyomma maculatum; States, although infection with H. americanum is diagnosed
for H. canis, the vector is Rhipicephalus sanguineus. Other much more frequently. H. canis has been identified in dogs
tick species (Amblyomma, Rhipicephalus, and Haemaphy- from Mississippi, Louisiana, Alabama, Georgia, Oklahoma, and
salis spp.) may also be involved. Virginia and in a dog that lived in New Jersey but was born in
a Texas shelter.16,17,29 A few dogs have mixed infections with
Major Clinical Signs: Dogs with H. americanum infections
both H. americanum and H. canis. Domestic cats can also be
most often show lethargy, fever, weight loss, locomo-
infected by Hepatozoon spp., which include H. felis and possibly
tory abnormalities, hyperesthesia, mucopurulent ocular
H. canis or closely related species (see later section on feline
discharge, signs related to protein-losing nephropathy
infections and Table 77-1).30,31
such as polyuria and polydipsia. Dogs with severe H. canis
Because dogs or other vertebrates serve as intermediate
infections may show fever and lethargy.
hosts, a vector is needed to complete the sexual phase of the life
Differential Diagnoses: Sterile or infectious meningitis, bacte- cycle of Hepatozoon species. In the case of H. americanum, the
rial discospondylitis, Lyme borreliosis, canine monocytic definitive host is the Gulf Coast tick, Amblyomma maculatum,
ehrlichiosis, toxoplasmosis, sarcocystosis, systemic immune- whereas the definitive host of H. canis is the brown dog tick,
mediated diseases such as systemic lupus erythematosus Rhipicephalus sanguineus.32,33 In Japan, Haemaphysalis spp.
Human Health Significance: Canine Hepatozoon species are ticks have been implicated as vectors of H. canis, and in Brazil,
not known to infect humans. Amblyomma ovale and Rhipicephalus (Boophilus) microplus
are suspected vectors.34-36
As it feeds on an infected dog, the nymphal tick ingests
circulating leukocytes that contain gamonts. Within the tick’s
Etiology and Epidemiology gut, the gamonts are freed from the cells and undergo further
development before fertilization occurs. The resulting zygote
Apicomplexan protozoal parasites from the genus Hepatozoon divides through sporogony and develops into an oocyst within
include more than 300 species that infect a wide variety of the hemocoel of the tick (Figure 77-2). Oocysts mature while the
amphibians, reptiles, birds, and mammals.3 Despite its name, tick molts to the adult stage. In the case of H. americanum, the
canine hepatozoonosis is not a zoonotic disease and rarely larval stage of the tick can also become infected, resulting in a
affects the liver. Canine hepatozoonosis is caused by one of two nymph that contains viable oocysts capable of transmitting the
species of Hepatozoon currently known to cause disease in dogs, disease.37 Each mature oocyst contains hundreds of sporocysts,
Hepatozoon canis and Hepatozoon americanum (compared in with each sporocyst containing 10 to 26 infective sporozoites.
Table 77-1). First described in India in 1905, H. canis has long Unlike most tick-borne diseases, hepatozoonosis is not transmit-
been known as the agent that infects dogs in many regions of ted through the bite of an infected tick but rather by ingestion of
the Old World, including Asia, Africa, southern Europe, and an infected tick. This typically occurs when the dog grooms itself
the Middle East, and more recently has been identified in the but can also occur if the dog feeds on tick-infested prey. Once
New World in both North and South America (Figure 77-1).1,3-20 ingested, exposure to bile in the dog’s intestinal tract results in
Hepatozoon americanum causes American canine hepatozo- release of the sporozoites, which penetrate the intestinal epithe-
onosis, which occurs in dogs primarily in the southeastern and lial wall and are transported to the target organs and tissues,
south-central United States. It was first confused with H. canis likely within mononuclear cells.

747
748 SECTION 4  Protozoal Diseases

TABLE 77-1
Comparison of Hepatozoon canis Infection, American Canine Hepatozoonosis, and Feline Hepatozoonosis
American Canine
H. canis Infection Hepatozoonosis Feline Hepatozoonosis
First described India, 1905 Texas Gulf Coast, 1978 India, 1908
Etiologic agent Hepatozoon canis Hepatozoon americanum Multiple poorly defined
Hepatozoon sp.
Primary vector Rhipicephalus sanguineus Amblyomma maculatum Unknown
(brown dog tick) (Gulf Coast tick)
Route of transmission Ingestion of tick Ingestion of tick Probably ingestion of tick
(not a tick bite) (not a tick bite) (not a tick bite)
Additional routes of Transplacental from dam to Ingestion of cystozoites Unknown
transmission puppy in transport hosts;
transplacental is likely
Affected mammalian hosts Dogs, foxes, jackals, hyenas, Dogs, coyotes, possibly bobcats, Domestic cats, wild felids
other carnivores ocelots, and other wildlife
Geographic distribution Africa, Middle East, Asia, Primarily southern and India, Africa, Israel, Brazil,
southern Europe, South southeastern United States Spain, France, Thailand,
America, United States United States (Hawaii)
Severity of disease Subclinical to mild; Usually severe, even in absence Typically subclinical unless
occasionally severe, esp. of immunosuppression immunosuppressed
if immunosuppressed
Major clinical signs Lethargy, fever, weight loss Fever, pain, lameness, ocular Nonspecific
discharge
Common Anemia Marked leukocytosis (20,000- Elevated CK activity
laboratory abnormalities Extreme leukocytosis is rare 200,000 cells/µL), anemia,
but may occur with high elevated ALP activity, low
parasitemia glucose concentration
Radiographic lesions Nonspecific; rare periostitis Common; periosteal bone Not reported
proliferation
Histopathology findings No muscle lesions Myositis, onion-skin cysts, Meronts in myocardium
“wheel-spoked” meronts pyogranulomas, and and skeletal muscle
in spleen, bone marrow, meronts in skeletal muscle
lymph nodes
Blood smear findings Gamonts common; parasitemia Rarely observed gamonts; Parasitemia is rare; when
of 1%-5% is typical; rarely parasitemia is usually present usually <1% of
see parasitemia of 5%-100% <0.1% neutrophils
Human health significance None known None known None known

In H. americanum infections, infected cells preferentially asexual reproductive cycle, or they may become gamonts. The
travel to skeletal muscle, where each organism develops within gamonts are then ingested by feeding ticks, which completes the
its host cell, which becomes lodged between myocytes (Figure life cycle. Some of the onion-skin cysts lie dormant for varying
77-3). Concentric layers of mucopolysaccharide are laid down lengths of time; their activation is responsible for the waxing
by the host cell to form an “onion-skin cyst,” which may protect and waning nature of the disease as well as the relapse of clinical
the organism from the immune response (Figure 77-4). Merogony signs that can occur months after perceived clinical cure.
occurs within the cyst, and on maturation of the meront, the In H. canis infections, infected cells are carried by lymph
cyst ruptures, and merozoites are released. This elicits a severe or blood to the spleen, bone marrow, lymph nodes, and other
inflammatory response, with recruitment of neutrophils and organs such as the liver, kidney, and lungs where the organisms
monocytes to the area. A pyogranuloma develops in the space divide asexually through merogony (Figure 77-6). Two types
where the cyst existed (Figure 77-5). Many of these inflammatory of meronts form: the “wheel spoke” meront, which contains
cells become infected with a single zoite. Angiogenesis within the 20 to 30 micromerozoites which form around a central round
pyogranuloma results in a highly vascular structure from which structure (Figure 77-7), and a meront that contains up to four
the infected cells can reenter circulation. The intracellular para- macromerozoites.38 Once released from the mature meront,
sites can travel to other target sites, where they continue the micromerozoites invade neutrophils and monocytes and become
CHAPTER 77  Canine and Feline Hepatozoonosis 749

FIGURE 77-1  Geographic distribution of reported Hepatozoon canis and Hepatozoon americanum infections. Both species exist in the United States. (Blue, Hepatozoon canis; red,
Hepatozoon americanum; purple both species.)

Other modes of transmission also occur in hepatozoonosis.


Transplacental transmission from dam to puppies has been doc-
umented for H. canis infections and likely occurs in H. america-
num infections as well.40 Ingestion of tissue from wild animals
or prey may transmit H. americanum.41-43 Laboratory rodents
fed H. americanum oocysts developed cystozoites, but they
did not develop gamonts or meronts or become ill. Cystozoite-
laden rodent tissue fed to naïve dogs resulted in H. americanum
infection along with its classical clinical signs. Investigation of
a natural outbreak in a pack of hunting beagles revealed that
clinical signs of American canine hepatozoonosis arose 4 to
6 weeks after some of the dogs were allowed to consume a wild
rabbit carcass, whereas dogs not allowed to consume the rabbit
did not develop clinical signs. The predation route of transmis-
sion has not yet been proven to occur with H. canis.
The geographic distribution of American canine hepato-
zoonosis aligns closely with the range of the Gulf Coast tick.
Although this tick is endemic in the states that surround the Gulf
FIGURE 77-2  Hepatozoon americanum oocysts from the hemocoel of an Amblyomma of Mexico, its range is expanding and it has been found as far
maculatum tick. Hundreds of small, round sporocysts are present within each oocyst. Each north as Kansas and Kentucky.44 Larval and nymphal stages of
sporocyst contains 10 to 26 infective sporozoites. A. maculatum preferentially feed on birds and small rodents.
Surveys of wildlife have revealed the presence of Hepatozoon
spp. that are related but not identical to H. americanum in small
rodents and rabbits.45 Although a reservoir species has not yet
gamonts, which can then be ingested by feeding ticks (Figure been identified, dogs are likely an aberrant, rather than natural,
77-8). The larger macromerozoites are believed to be responsible host of H. americanum. H. americanum or closely related species
for the production of secondary meronts in the target tissues, have been identified in coyotes, bobcats, and ocelots.46-49 Because
which continue the asexual cycle of merogony. Another tissue these wild animals were in good physical condition at the time
stage found in dogs with H. canis but not H. americanum infec- of capture, they may represent a reservoir host in nature. In con-
tions is a small monozoic cyst that resembles the cystozoite found trast to adult animals, coyote puppies developed classic signs of
in transport hosts of other Hepatozoon species.39 H. americanum infection after experimental transmission.50
750 SECTION 4  Protozoal Diseases

FIGURE 77-5  Pyogranuloma in skeletal muscle of a dog infected with Hepatozoon


americanum. Zoites displace the nucleus in several of the inflammatory cells.

There is no sex or breed predilection for hepatozoonosis, but


hunting dogs, rural dogs, and dogs allowed to roam are most
at risk.

Clinical Features
Signs and Their Pathogenesis
Dogs with H. americanum infection exhibit a moderate to
FIGURE 77-3  Life cycle of Hepatozoon americanum. severe illness, whereas most dogs infected with H. canis have no
or only mild signs of illness. Immunosuppression or concurrent
disease is an important factor in expression of illness in H. canis
but not in H. americanum infections. Co-infections with other
pathogens such as Ehrlichia, Babesia, Anaplasma, Leishmania,
parvovirus, or canine distemper virus can allow establishment
of a new H. canis infection as well as progression or reactivation
of an existing infection.
Clinical signs of H. americanum infection are associated
with the strong inflammatory response that occurs when
meronts rupture, leukocytes are recruited, and pyogranu-
lomas form in skeletal muscle. The earliest lesions occur
3 weeks after infection.51 As the organisms multiply, the
infection disseminates, which results in more severe inflam-
mation that waxes and wanes over time. The inflammation
causes fever and myositis, which is associated with locomo-
tor abnormalities and hyperesthesia (Table 77-2). The clini-
cal signs can mimic those of meningitis or discospondylitis.
Many affected dogs also develop bone lesions that are evi-
dent radiographically and resemble lesions seen in hypertro-
phic osteopathy, except they are proximal rather than distal
in distribution (Figure 77-9). It is unknown whether the bone
lesions result from nearby localized muscle inflammation or
FIGURE 77-4  Skeletal muscle containing a developing meront of H. americanum. from humoral factors. Dogs usually have a mucopurulent
The mucopolysaccharide layers produced by the host cell protect the developing organ- ocular discharge that may be caused by pyogranulomatous
isms from the host’s immune system. inflammation of the extraocular muscles or of the lacri-
mal gland (Figure 77-10). Return of ocular discharge is fre-
Dogs are believed to be the natural host reservoir of H. canis. quently the first indication of a relapse following treatment.
The brown dog tick, which serves as the definitive host of Dogs with American canine hepatozoonosis frequently main-
H. canis, is found worldwide, and all three stages preferentially tain a fairly normal appetite; however, weight loss, cachexia,
feed on dogs. Life stages of H. canis or morphologically indis- and muscle atrophy occur over time. There may be a history
tinguishable Hepatozoon spp. have been reported in numerous of polyuria and polydipsia that is due to glomerulonephritis
carnivore species around the world, including several species of or amyloidosis, which can occur secondary to long-standing
fox, jackal, African wild dog, hyena, palm civet, cheetah, leopard, inflammation. Nephrotic syndrome and thromboembolic
lion, and Pallas cat.30 complications may ensue. Less common clinical signs include
CHAPTER 77  Canine and Feline Hepatozoonosis 751

FIGURE 77-6  Life cycle of Hepatozoon canis.

FIGURE 77-7  Hepatozoon canis meront in splenic tissue of a dog from Israel dem-
onstrating the typical “wheel spoke” pattern. (Courtesy of Dr. Gad Baneth, Koret School of
Veterinary Medicine, Israel.)
FIGURE 77-8  Two Hepatozoon canis gamonts in neutrophils on a blood smear. The
gamonts of Hepatozoon americanum are nearly identical in appearance to those of H. canis,
although they are rarely seen. 1000x magnification. (Courtesy of Dr. Gad Baneth, Koret
diarrhea, mucosal pallor, cough, abnormal lung sounds, and School of Veterinary Medicine, Israel.)
lymphadenomegaly.
Most dogs infected with H. canis have a low level of parasit-
emia, with less than 5% of circulating leukocytes infected with of the disease, body temperature may be normal at any given
gamonts.52 These dogs have a limited inflammatory reaction. time.23 Because of the myositis, dogs are typically evaluated for
About 15% of parasitemic dogs have a high level of parasitemia gait abnormalities, which range from lameness or stiffness to
(>800 gamonts/µL). The degree of parasitemia correlates with recumbency and an inability to rise. Hyperesthesia is common
the severity of clinical signs; the sickest dogs have a level of and may appear as cervical, back, joint, or generalized pain.
parasitemia that approaches 100% of infected circulating leu- Lethargy is common, and cachexia and generalized muscle atro-
kocytes. These dogs may have hepatitis, glomerulonephritis, or phy may be apparent. Mucopurulent ocular discharge is often
pneumonitis in addition to severe anemia, fever, and cachexia. present. Other ocular lesions seen on occasion include focal
retinal scars or hyper-reflectivity, increased retinal pigmenta-
Physical Examination Findings tion, papilledema, and uveitis with active inflammatory fundic
With H. americanum infection, fever (up to 105.6°F or 40.9°C) lesions. Mucosal pallor and lymphadenomegaly may also be
is common, although because of the waxing and waning nature present.
752 SECTION 4  Protozoal Diseases

TABLE 77-2
Frequency of Clinical Signs in 22 Dogs with American Canine
Hepatozoonosis
Clinical Sign Number of Dogs (%)
Fever 19 (86)
Weight loss 18 (82)
Mucopurulent ocular discharge 17 (77)
Low tear production 8 (36)
Muscle atrophy 14 (64)
Pain (all types) 14 (64)
Joints 2 (9)
Lumbar 4 (18)
Cervical 5 (23)
Generalized 3 (14)
Stiffness 12 (55)
Generalized weakness 9 (41)
Pelvic limb paresis and ataxia 5 (23)
Inability to rise 5 (23)
Anorexia 5 (23)
FIGURE 77-9  Radiograph of the pelvic limb of a dog with Hepatozoon americanum
From Vincent-Johnson NA. American canine hepatozoonosis. Vet Clin infection. There is smooth periosteal proliferation on the cranial aspect of the femur.
North Am Small Anim Pract. 2003;33:905-920. Data from Macintire (From Vincent-Johnson NA. American canine hepatozoonosis. Vet Clin North Am Small
DK, Vincent-Johnson N, Dillon AR, et al. Hepatozoonosis in dogs: 22 Anim Pract. 2003;33:905-920.)
cases (1989-1994). J Am Vet Med Assoc. 1997;210:916-922.

Dogs with mild H. canis infections may have pale mucous


membranes and lethargy. Dogs with high parasitemia often
exhibit fever, lethargy, and severe weight loss or cachexia, even if
they maintain a good appetite. Splenomegaly and lymphadeno-
megaly may be detected. Dogs often have physical examination
abnormalities that relate to the presence of co-infections or other
comorbidities.

Diagnosis
Laboratory Abnormalities
Complete Blood Count
The most common laboratory abnormality in H. americanum
infection is leukocytosis, which is often extreme (Table 77-3).
White cell counts are typically 20,000 to 200,000 cells/µL,
with reported means of 76,807 and 85,700 cells/µL.23,53 The
leukocytosis is due to a mature neutrophilia, although some-
times there is a mild to moderate left shift. A mild normocytic,
normochromic nonregenerative anemia is typical. Platelets are
usually normal to increased, sometimes with thrombocytosis
up to 916,000 platelets/µL. When thrombocytopenia is evident,
concurrent tick-borne diseases such as ehrlichiosis should be
considered. FIGURE 77-10  Rottweiler with Hepatozoon americanum infection. Note the hunched
The most common laboratory finding in H. canis infec- appearance, muscle atrophy, and mucopurulent ocular discharge.
tion is anemia, which is usually normocytic, normochromic,
and occasionally regenerative.52 Platelets are decreased about
one third of the time, but this may be due to concurrent infec- Serum Chemistry Profile
tion with Ehrlichia canis or other disease. The white blood cell In H. americanum infection, a mild elevation in the activity
count is typically normal in cases with low parasitemia but is of ALP is typical, possibly due to periosteal new bone forma-
elevated in dogs with high parasitemia. Some dogs have neu- tion. Serum glucose concentration is often decreased (40 to
trophil counts of 50,000 to 150,000/µL with close to 100% of 60 mg/dL) and occasionally as low as 5 mg/dL. This is not
these cells containing gamonts. a true hypoglycemia, but rather a laboratory artifact due to
CHAPTER 77  Canine and Feline Hepatozoonosis 753

new bone formation occurs most frequently and severely on


TABLE 77-3 the diaphysis of long bones but occurs to a lesser degree on
Frequency of Laboratory and Radiographic Findings in 22 Dogs flat and irregular bones. The radiographic appearance can vary
from subtle irregular periosteal exostoses to thick parallel pseu-
with American Canine Hepatozoonosis
docortices. H. americanum infection typically affects the more
proximal bones while sparing the distal bones.56
Finding Number of Dogs (%)
In H. canis infections, radiographic bone lesions are rare.
Moderate to marked leukocytosis* 22 (100) Periostitis has only been described in three reports from Japan,
Mature neutrophilia 15 (68) India, and Italy, and in an experimental infection in Israel.4,57
Mild left shift 7 (31)
Elevated ALP activity 22 (100) Microbiologic Testing
Hypoglycemia 20 (91) Diagnostic assays currently available for hepatozoonosis in
dogs are described in Table 77-4.
Hypoalbuminemia 19 (86)
Periosteal proliferation on 18 (82) Cytologic Diagnosis
radiography The sexual stage of H. americanum and H. canis, the gamont,
Anemia (nonregenerative) 14 (64) appears as a light blue to clear oblong structure with a faintly
staining nucleus within the cytoplasm of neutrophils or monocytes
Hypocalcemia 14 (64)
stained with Diff Quik or Giemsa. The gamonts of the two species
Normal platelet count 11 (50) look nearly identical, although there is a slight difference in size:
Thrombocytosis 9 (41) H. canis gamonts measure approximately 11.0 × 4.3 µm while
Hyperphosphatemia 7 (32) H. americanum gamonts are slightly smaller at 8.8 × 3.9 µm.26
In dogs with H. americanum infection, gamonts are rarely
Low BUN concentration 7 (32)
found within monocytes or neutrophils on peripheral blood
Hyperglobulinemia 4 (11) smears. When present, infected cells rarely exceed 0.1% of the
Hypercalcemia 2 (9) circulating leukocytes; several thousand cells may have to be
Elevated CK activity 1 (5) examined before an infected cell is found. The chance that gam-
onts are detected can be increased by examination of buffy-coat
From Vincent-Johnson NA. American canine hepatozoonosis. Vet Clin smears. Neither bone marrow nor lymph node aspirates are of
North Am Small Anim Pract. 2003;33:905-920. Data from Macintire much value in making a definitive diagnosis of H. americanum
DK, Vincent-Johnson N, Dillon AR, et al. Hepatozoonosis in dogs: 22 infection; organisms are not usually seen in these specimens.
cases (1989-1994). J Am Vet Med Assoc. 1997;210:916-922. In most dogs with H. canis infections, gamonts are found
*Minimum white cell count was 27,800 cells/µL. on blood smear examination in 0.5% to 5% of neutrophils and
monocytes. Parasitemia may approach 100% in heavy infec-
tions. Gamonts may also be identified in dogs that are apparently
utilization of glucose in the specimen by the high number of healthy.
white blood cells. Hypoalbuminemia is common and may be
due to chronic inflammation, decreased protein intake, or Serologic Diagnosis
renal loss from secondary glomerulonephritis or amyloidosis. An ELISA assay for detection of antibodies to H. americanum
Except in dogs with significant renal damage, BUN concentra- has been developed at Oklahoma State University for research
tion is often low. The combination of increased activity of purposes, but this assay is not available commercially to veteri-
serum ALP and low glucose, BUN, and albumin concentra- nary practitioners.58 Both an indirect immunofluorescent anti-
tions may mislead the clinician to suspect liver disease. Serum body (IFA) assay and ELISA assays for H. canis antibodies have
bile acid concentrations are normal or only slightly elevated. been developed and used for epidemiologic studies in countries
Surprisingly, the activity of serum CK is consistently within outside of the United States.59-62 Positive serologic test results
the normal range even though H. americanum infection causes indicate exposure but do not prove active infection or correlate
severe myositis. with clinical disease. Thus, serologic testing is mostly of use for
Common serum chemistry findings in H. canis infection epidemiologic studies and for screening blood donor animals.
include hyperproteinemia, hyperglobulinemia, and hypoalbu-
minemia. The hyperglobulinemia is due to a polyclonal gam- Molecular Diagnosis Using the Polymerase Chain Reaction
mopathy.52,54 Increases in the activities of ALP and CK are A real-time PCR assay that detects Hepatozoon spp. is available
usually present. commercially to veterinary practitioners through the Auburn
University Molecular Diagnostics Laboratory.63 Other com-
Diagnostic Imaging mercial veterinary diagnostic laboratories also offer real-time
Plain Radiography PCR assays for detection of Hepatozoon species. The assay per-
Many dogs with H. americanum infection show radiographic formed at Auburn University detects as few as seven genomic
abnormalities of the skeletal system; therefore, radiographs of copies of Hepatozoon spp. per mL of blood, and it distinguishes
the pelvis or long bones can be useful to support a clinical diag- between infection with H. canis and H. americanum.16,17
nosis of American canine hepatozoonosis. Young dogs are more Although highly sensitive and specific, false negatives can occur
likely than older dogs to show bone lesions. In experimental early in the course of infection or in dogs with chronic disease
infections, bone lesions are recognizable histologically within where the numbers of circulating gamonts are extremely low or
5 weeks postexposure.55 Disseminated, symmetric periosteal absent. Muscle biopsy should be performed in dogs suspected
754 SECTION 4  Protozoal Diseases

TABLE 77-4
Diagnostic Assays Currently Available for Hepatozoonosis in Dogs
Assay Specimen Type Target Performance
Cytologic examination Whole blood smears Hepatozoon spp. False negatives are common because of low circulat-
gamonts in leukocytes ing numbers of gamonts, especially in clinical
H. americanum infections.
Antibody serology Serum Antibodies to H. canis Positive test results indicate exposure but do not
(ELISA or immuno­ indicate the presence of the organism in circulat-
fluorescent antibody) ing leukocytes and do not correlate with clinical
disease.
Real-time PCR assays Whole blood H. americanum or Sensitivity and specificity may vary depending on
H. canis DNA assay design, but in general PCR assays are more
sensitive than cytology. False-negative results may
occur when organism levels are very low, such as
early in disease or in very chronic infections.
Histopathology Muscle biopsy H. americanum cysts, Most reliable method of diagnosis of H. americanum
specimens meronts, and pyogran- infection, but requires anesthesia.
ulomas

to have H. americanum infection but that have negative PCR macrophages, many of which contain a single zoite. Myositis is
assay results. characterized by muscle necrosis and atrophy with infiltration of
neutrophils, macrophages, and occasional lymphocytes between
Pathologic Findings muscle fibers. Although consistently identified in skeletal and car-
Gross Pathologic Findings diac muscle, H. americanum cysts, meronts, and pyogranulomas
In dogs with H. americanum infection, muscle atrophy and can sporadically be found in adipose tissue, intestinal smooth
cachexia are consistent findings on gross necropsy examination. muscle, pancreas, liver, lymph node, spleen, skin, salivary gland,
Pyogranulomas may appear as multiple 1- to 2-mm–diameter lung, and kidney.64 Renal damage is common and manifests as
white-tan foci.23 These are scattered diffusely throughout skeletal focal pyogranulomatous inflammation with mild glomerulone-
and cardiac muscle but may also be observed in other tissues. phritis, lymphoplasmacytic interstitial nephritis, mesangioprolif-
Roughening and thickening of bony surfaces are often present. erative glomerulonephritis, or occasionally amyloidosis. Amyloid
Less common findings include lymphadenomegaly, congestion deposits have also been found in the spleen, lymph nodes, small
of the gastric mucosa, splenic coagulative necrosis, and pulmonary intestines, and liver of affected dogs. Various organs can show
congestion. vascular changes, which include fibrinoid degeneration of ves-
In dogs with H. canis infection, the spectrum of gross patho- sel walls, mineralization and proliferation of the vascular intima,
logic lesions depends on the severity of the infection. In heavy and pyogranulomatous vasculitis.
infections, splenomegaly and hepatomegaly are common findings In H. canis infections, “wheel-spoked” meronts are present in
and these organs often exhibit diffuse white necrotic foci that are varying numbers and stages of development throughout hemo-
1 to 2 mm in diameter.14 The necrotic areas may be larger and lymphatic target organs, such as bone marrow, spleen, and
nodular and can also be found in other tissues, such as pancreas lymph nodes. The meronts are approximately 30 µm in diameter
and pleura. Lymphadenomegaly is common, and pneumonia and on central cross-section appear as a circle of micromerozoite
may be present. nuclei surrounding a central core. Meronts can be found in small
numbers as an incidental finding in dogs from endemic areas.65
Histopathologic Findings Inflammation associated with meronts can range from none to
For H. americanum infections, muscle biopsy has long been con- very severe. In heavy infections, microscopic lesions may include
sidered the most reliable method for diagnosis. Small pieces of hepatitis with Kupffer’s cell hyperplasia and inflammatory infil-
muscle (approximately 2 cm × 2 cm) are obtained from the biceps trates, interstitial pneumonia and thickening of alveolar septae
femoris or semitendinosus muscles under general anesthesia.25 with inflammatory infiltrates, glomerulonephritis or interstitial
Submission of multiple specimens increases sensitivity, especially nephritis with multifocal necrosis, and focal splenic necrosis.
in low-level infections. Muscle lesions consist of large onion-skin
cysts, pyogranulomas, and myositis. The cysts are round to oval Treatment and Prognosis
and have mean cross-section dimensions of 186 × 150 µm.26 At
the center of the cyst is a host cell nucleus and protozoa, but Although no treatment effectively eliminates the tissue stages
depending on the plane of the cut surface, these may or may not of H. americanum, remission of clinical signs can be achieved
be visible. Surrounding the center of the cyst are concentric layers using a combination of a trimethoprim-sulfa, clindamycin,
of mucopolysaccharide. Occasionally meronts in various stages and pyrimethamine for 14 days (Table 77-5).66 Response to
of development are observed within these cystic structures. The therapy is usually dramatic, with resolution of clinical abnor-
highly vascular pyogranulomas are packed with neutrophils and malities within 48 to 72 hours of initiating therapy. Ponazuril
CHAPTER 77  Canine and Feline Hepatozoonosis 755

TABLE 77-5
Antimicrobial Drugs Indicated for Treatment of American Canine Hepatozoonosis

Drug Dose* (mg/kg) Route Interval (hours) Duration (days)


TCP combination:
Trimethoprim-sulfonamide and 15 PO 12 14
Clindamycin and 10 PO 8 14
Pyrimethamine and 0.25 PO 24 14
Decoquinate† 10-20 PO 12 730

Adapted from Baneth G, Macintire DK, Vincent-Johnson N, et al. Hepatozoonosis. In Greene CE, ed. Infectious Diseases of the Dog and Cat, 3 ed.
St. Louis, MO: Saunders; 2006:698-711.
TCP, Trimethoprim-sulfadiazine, clindamycin, and pyrimethamine; PO, by mouth.
*Dose per administration at specified interval.
†Use the decoquinate 6% (27.2 g/lb) powder, Deccox (Alpharma Inc., Fort Lee, NJ). One teaspoonful is equivalent to approximately 180 mg

decoquinate. Administer at a rate of 1 teaspoon per 10 kg of body weight and feed q12h mixed into moist dog food.

(10 mg/kg PO q12h for 14 days) is an alternative therapeutic imidocarb therapy.69 Despite lack of a parasitologic cure, clini-
option.67 Either of these protocols must be followed by long- cal cure is achieved in many dogs. Dogs with low parasitemia
term administration of decoquinate in order to prevent relapse. generally have a good long-term prognosis for survival, whereas
Without the addition of decoquinate, relapse occurs in most those with high parasitemia have a guarded prognosis.
dogs 2 to 6 months following treatment. Although these dogs
generally respond well to another round of combination ther- Immunity and Vaccination
apy, subsequent relapses occur more frequently, and eventually
disease becomes refractory to treatment. Persistent infections Current treatments do not cure H. americanum or H. canis
and multiple relapses lead to complications such as glomeru- infections, so dogs remain susceptible to relapse of disease, espe-
lonephropathy, amyloidosis, vasculitis, and cachexia, which cially if immunosuppressed. In experimental H. canis infections,
carry a guarded to poor prognosis. Decoquinate, a livestock treatment with an immunosuppressive dose of glucocorticoids
anticoccidial agent, apparently breaks the asexual recycling was followed by the appearance of parasitemia.33 Vaccines for
of infection by arresting development of the merozoites after H. americanum or H. canis infections are not available.
their release from meronts. Two years of daily decoquinate
therapy is recommended.66 Alternatively, a PCR assay can be Prevention
performed every 3 to 6 months, and when the PCR becomes
negative, treatment can be discontinued. Mild relapses can Since both H. canis and H. americanum are transmitted by
occur even during treatment with decoquinate. If clinical signs ingestion of ticks, the use of topical acaricides, environmental
are severe, another course of combination therapy is indicated. parasiticides, and immediate removal of ticks from dogs are the
Long-term administration of decoquinate results in extended most important means of prevention. Because H. americanum,
survival times and excellent quality of life with a good prog- and possibly H. canis, can also be transmitted through ingestion
nosis. During the initial few days of treatment with either of cystozoites in muscle from other animals, dogs should not be
antiprotozoal regimen, administration of a nonsteroidal anti- allowed to roam, scavenge, or engage in predatory behavior.
inflammatory drug at standard dosages can provide relief from Dogs should not be fed uncooked or undercooked game meat.
fever and pain. Although Hepatozoon spp. have not been shown to be transmitted
Hepatozoon canis infections are treated with imidocarb through blood transfusion, it seems logical that dogs infected
dipropionate (5 to 6 mg/kg SC or IM every 14 days) until with Hepatozoon species be avoided as blood donor dogs. To
gamonts are no longer seen on blood smear examination.52 prevent congenital infections of puppies, infected females either
Although some dogs require only one or two treatments, at should not be bred or should receive treatment before breeding.
least 8 weeks of treatment may be required for dogs with heavy
infections. Doxycycline (10 mg/kg/day PO for 21 to 28 days) is Public Health Aspects
frequently given in conjunction with imidocarb dipropionate.
It has been recommended that all dogs, even those with only Only one case of human infection with a Hepatozoon spp. has
mild disease, be treated, because parasitemia may increase over been reported. Gamonts were identified on blood smears of
time. A recent study showed that despite negative blood smears, an adult man from the Philippines who suffered from anemia
dogs remained positive by PCR or buffy-coat examination even and icterus, but no parasites were found in liver or bone mar-
when treated repeatedly over 8 months.68 This study also found row biopsies.70 Because hepatozoonosis is transmitted by tick
that buffy-coat examination was twice as sensitive as blood ingestion rather than a tick bite, tick ingestion is an unlikely
smear examination in detecting gamonts, and PCR was even route of transmission for humans. However, caution should
more sensitive for detection of persistent infection. Addition of be used when removing ticks from dogs or when handling
toltrazuril does not seem to provide any additional benefit to tick-infested dogs or blood from infected dogs because of the
756 SECTION 4  Protozoal Diseases

unknown zoonotic potential of organisms that cause canine smears.7 In Israel, Hepatozoon DNA was amplified from the
disease and the possibility that known human pathogens may blood of 36% of 152 cats, and infected cats were significantly
also be present. more likely to have access to the outdoors.31 The vast majority
of cats were infected with an organism that resembled H. felis,
Hepatozoonosis in Domestic Cats although 2 cats were infected with an organism that resembled
H. canis. Most infections were subclinical. Meronts have also
Feline hepatozoonosis occurs primarily in areas where canine been found in the myocardium and skeletal muscles of cats
hepatozoonosis exists, and it has been reported in a number with no signs of illness. Cats with clinical hepatozoonosis are
of countries, including India, South Africa, Nigeria, Brazil, commonly immunosuppressed because of infection with FIV
Israel, Spain, France, and Thailand.30,31 In the United States, or FeLV. A range of clinical signs has been reported, including
there is one report of hepatozoonosis in a domestic cat from weakness, intermittent anorexia, weight loss, fever, lymph-
Hawaii.71 A survey of blood specimens from stray cats in adenomegaly, ulcerative glossitis, hypersalivation, anemia,
Bangkok, Thailand, showed that 32.3% were positive by serous ocular discharge, and icterus. Treatment success has
PCR for a Hepatozoon sp. most closely related to H. canis, been reported with oral doxycycline at 5 mg/kg PO once a day
whereas only 0.7% of the cats had gamonts observed on blood for 10 days.72

CASE EXAMPLE
Signalment: “Pepper,” a 1.5-year-old male miniature schnauzer
from Opelika, AL (Figure 77-11)
History: Pepper was seen for a 1-month duration of illness
characterized by intermittent fever (104.0°F to 105.0°F [40.0°C
to 40.6°C]), lethargy, and ocular discharge. His owner reported
that Pepper had also been stiff and reluctant to move. Despite
a fairly good appetite, he had lost weight and muscle mass.
The fever and other clinical signs had been nonresponsive
to antibiotics, which included enrofloxacin and doxycycline,
prescribed at the onset of illness. Pepper was a house dog
but spent time outdoors in a rural environment. No vomiting,
coughing, or sneezing had been reported, but he had had
mild diarrhea at the onset of illness that resolved after a week.
He had also had a mild increase in thirst and urination. He was FIGURE 77-11  “Pepper,” a 1.5-year-old male miniature schnauzer with H.
fed a high-quality commercial dry dog food, received monthly americanum infection.
heartworm preventive, and was current on all vaccinations,
including canine distemper, hepatitis, parvovirus, and rabies Ophthalmic Exam: Pupillary light reflex (direct and
vaccines. He had not traveled outside of the local area. The consensual): Normal.
owner mentioned that she had had another dog that died Conjunctiva: injected bilaterally, mucopurulent discharge bilat-
about 1 year earlier after exhibiting similar clinical signs, but a erally.
diagnosis was not made. Cornea and lens: Normal.
Current medications: Aspirin 11 mg/kg PO q12h for pain. Schirmer Tear Test: Right eye = 18 mm; left eye = 16 mm (normal
Physical Examination: > 15 mm).
Body Weight: 7.6 kg. Fluorescein stain: Negative.
General: Quiet, slightly lethargic, but responsive. Reluctant to Tonometry: Normal.
walk or stand; appeared painful. T = 103.8°F (39.9°C), HR = 96 Fundoscopic examination: A small grayish, slightly raised focal
beats/min, RR = 32 breaths/min, mucous membranes pink, lesion was identified in the right fundus.
CRT = 1.5 s. Laboratory Findings:
Integument: The skin and haircoat appeared normal with no CBC:
evidence of ectoparasites. PCV 38.7% (37%-55%)
Eyes, Ears, Nose, and Mouth: A moderate amount of MCV 68.7 fL (60-77 fL)
mucopurulent ocular discharge was present bilaterally. No MCHC 35.4 g/dL (32-36 g/dL)
other abnormalities were identified. WBC 39,200 cells/µL (6000-17,000 cells/µL)
Musculoskeletal: Body condition score was 3/9 with Neutrophils 35,672 cells/µL (3000-11,400 cells/µL)
moderate generalized muscle atrophy. The dog exhibited Lymphocytes 1176 cells/µL (1000-4000 cells/µL)
hyperesthesia during palpation of trunk, head, neck, and Monocytes 2352 cells/µL (150-1200 cells/µL)
limb muscles. The dog’s gait was very stiff and slow. Eosinophils 0 (100-750 cells/µL)
All Other Systems: Apart from abdominal splinting on Platelets 607,000 platelets/µL (200,000-400,000 platelets/µL).
palpation of the abdomen, no clinically significant The clinical pathologist’s blood smear review showed no
abnormalities were present. evidence of microorganisms.
CHAPTER 77  Canine and Feline Hepatozoonosis 757

Serum Chemistry Profile: the cytoplasm of <0.1% of the leukocytes. These were
BUN 8 mg/dL (10-25 mg/dL) characteristic of Hepatozoon gamonts.
Creatinine 0.6 mg/dL (0.3-1.0 mg/dL) Diagnosis: American canine hepatozoonosis (Hepatozoon
ALP 142 U/L (19-50 U/L) americanum infection).
ALT 37 U/L (17-66 U/L) Treatment: Triple combination therapy consisting of
Creatine kinase 148 U/L (92-357 U/L) (1) pyrimethamine 0.5 mg/kg PO q24h for 14 days;
Total bilirubin 0.3 mg/dL (0.1-0.3 mg/dL) (2) sulfadiazine/trimethoprim 15 mg/kg PO q12h for
Glucose 52 mg/dL (80-100 mg/dL) 14 days; and (3) clindamycin 10 mg/kg PO q8h for 14 days.
Sodium 153 mmol/L (146-160 mmol/L) This was associated with resolution of fever within
Potassium 4.7 mmol/L (3.5-5.9 mmol/L) 24 hours. According to his owner, Pepper was back to normal
Chloride 118 mmol/L (108-125 mmol/L) within 7 days. After the 14-day course of triple combination
Calcium 10.3 mg/dL (9.5-11.8 mg/dL) therapy was completed, Pepper was started on decoquinate
Phosphorus 3.9 mg/dL (3.3-5.8 mg/dL) 15 mg/kg PO q12h with food. At day 74, the owners ran out of
Total protein 6.4 g/dL (5.1-7.3 g/dL) decoquinate and Pepper missed 2 days of treatment before a
Albumin 2.7 g/dL (2.6-3.5 g/dL) refill was obtained. Eight days later, the owners called to say
Globulin 3.7 g/dL (3.6-5.0 g/dL) that Pepper was doing poorly, could not get up, and his eyes
Total CO2 21.8 mmol/L (13.9-31.5 mmol/L). were matted shut with a yellow-green discharge. Physical
Urinalysis: SGr 1.035; pH 8.0, protein 1+, bilirubin negative, exam and CBC findings were similar to initial findings.
hemoprotein negative, glucose negative, 0 WBC/HPF, 0 RBC/ Another 14-day course of triple combination therapy with
HPF, no crystals, moderate lipid droplets. pyrimethamine, sulfadiazine/trimethoprim, and clindamycin
Imaging Findings: was prescribed. Once again, Pepper’s clinical signs resolved
Pelvic Radiographs: Bilateral smooth periosteal bone within a few days. Pepper has continued on the decoquinate
formation was present on the ilium and femurs. twice daily with no further breaks in doses and no more
Abdominal Ultrasound Examination: No abnormalities were relapses.
detected. Comments: American canine hepatozoonosis was suspected
Bone Marrow Aspirate: The bone marrow smears were in this dog because of the geographic location in an endemic
highly cellular with many particles. Megakaryocytes were area of the southeastern United States, a waxing and waning
adequate. In a 500-cell count, the M:E ratio was 1.7 (reference fever, muscle pain, ocular discharge, leukocytosis, and
range 1.3-2.1) with normal maturation sequences in both other laboratory findings. As initially occurred in this case,
red and white series. Multiple smears examined showed no organisms often cannot be seen on blood smear or buffy-
evidence of microorganisms. coat exam. Muscle biopsy has been considered the most
Muscle Biopsy: Histopathology showed infiltration of reliable way to diagnose infection, although PCR is also a
neutrophils and monocytes between muscle fibers. There very sensitive and specific diagnostic tool. This case illustrates
were rare onion-skin cysts and one focal pyogranuloma. the typical relapsing nature of the disease and the need for
Many of the monocytes and neutrophils within the continuous long-term decoquinate therapy. After missing
granuloma contained a single zoite. Findings were consistent only a few doses of decoquinate, this dog suffered a relapse
with Hepatozoon americanum infection. days later. He made a quick recovery with a second round of
Cytology Findings: Although negative at the initial triple combination therapy. Effective treatment requires daily
evaluation, blood smears and buffy-coat smears performed decoquinate therapy with no breaks for at least 2 years or
a few days later showed oblong “jellybean” inclusions in until PCR of whole blood becomes negative.

SUGGESTED READINGS 5. Rajamanickam C, Wiesenhutter E, Zin FMD, et al. The incidence


of canine haematozoa in peninsular Malaysia. Vet Parasitol.
Baneth G. Perspectives on canine and feline hepatozoonosis. Vet Para- 1985;17:151-157.
sitol. 2011;181:3-11. 6. Novilla MN, Kwapien RP, Peneyra RS. Occurrence of canine
Little SE, Allen KE, Johnson EM, et  al. New developments in canine hepatozoonosis in the Philippines. Proc Helminthol Soc Wash.
hepatozoonosis in North America: a review. 4th International Canine 1977;44:98-101.
Vector-Borne Disease Symposium, Seville, Spain, 26-28 March 2009. 7. Jittapalapong S, Rungphisutthipongse O, Maruyama S, et  al.
Parasites Vectors. 2009;2(Suppl I):S5. Detection of Hepatozoon canis in stray dogs and cats in Bangkok,
Vincent-Johnson NA. American canine hepatozoonosis. Vet Clin North Thailand. Ann N Y Acad Sci. 2006;1081:479-488.
Am Small Anim Pract. 2003;33:905-920. 8. McCully RM, Basson PA, Bigalke RD, et  al. Observations
on naturally acquired hepatozoonosis of wild carnivores and
REFERENCES dogs in the Republic of South Africa. Onderstepoort J Vet Res.
1. James SP. On a parasite found in the white corpuscles of the blood 1975;42:117-134.
of dogs. Sci Mem Offrs Med Sanit Deps India. 1905;14:1-12. 9. Ezeokoli CD, Ogunkoya AB, Abdullah R, et al. Clinical and epi-
2. Craig TM, Smallwood JE, Knauer KW, et  al. Hepatozoon canis demiological studies on canine hepatozoonosis in Zaria, Nigeria.
infection in dogs: clinical, radiographic, and hematologic findings. J Small Anim Pract. 1983;24:455-460.
J Am Vet Med Assoc. 1978;173(8):967-972. 10. Oyamada M, Davoust B, Boni M, et  al. H. Detection of Babesia
3. Smith TG. The genus Hepatozoon. J Parasitol. 1996;82:565-585. canis rossi, B. canis vogeli, and Hepatozoon canis in dogs in a village
4. Murata T, Shiramizu K, Hara Y, et al. First case of Hepatozoon canis of eastern Sudan by using a screening PCR and sequencing method-
infection of a dog in Japan. J Vet Med Sci. 1991;53:1097-1099. ologies. Clin Diagn Lab Immunol. 2005;12:1343-2136.
758 SECTION 4  Protozoal Diseases

11. Kontos V, Koutinas A. Canine hepatozoonosis: a review of 11 nat- 34. Murata T, Inoue M, Taura Y, et al. Detection of Hepatozoon canis
urally occurring cases. Bull Hell Vet Med Soc. 1990;41:73-81. oocyst from ticks collected from the infected dogs. J Vet Med Sci.
12. Fischer S, Hartmann K, Gothe R. Hepatozoon canis: eine importi- 1995;57:111-112.
erte parasitare Infektion bei Hunden. Tierarztl Prax. 1994;22: 35. Forlano M, Scofield A, Elisei C, et  al. Diagnosis of Hepatozoon
172-180. spp. in Amblyomma ovale and its experimental transmission in
13. Hervas J, Carrasco L, Gomez-Villamanddos JC, et al. Acute fatal domestic dogs in Brazil. Vet Parasitol. 2005;134:1-7.
hepatozoonosis in a puppy: histopathological and ultrastructural 36. de Miranda RL, de Castro JR, Olegario MM, et al. Oocysts of Hep-
study. Vet Rec. 1995;137:518-519. atozoon canis in Rhipicephalus (Boophilus) microplus collected
14. Baneth G, Harmelin A, Presentey BZ. Hepatozoon canis in two from a naturally infected dog. Vet Parasitol. 2011;177:392-396.
dogs. J Am Vet Med Assoc. 1995;206:1891-1894. 37. Ewing SA, DuBois JG, Mathew JS, et al. Larval Gulf Coast ticks
15. Elias E, Homans PA. Hepatozoon canis infection in dogs: clini- (Amblyomma maculatum) [Acari: Ixodidae] as host for Hepa-
cal and hematological findings; treatment. J Small Anim Pract. tozoon americanum [Apicomplexa: Adeleorina]. Vet Parasitol.
1988;29:55-62. 2002;103:43-51.
16. Allen LE, Li Y, Kaltenboeck B, et al. Diversity of Hepatozoon species 38. Baneth G, Samish M, Shkap V. Life cycle of Hepatozoon canis
in naturally infected dogs in the southern United States. Vet Parasitol. (Apicomplexa: Adeleorina: hepatozoidae) in the tick Rhipicepha-
2008;154:220-225. lus sanguineus and domestic dog (Canis familiaris). J Parasitol.
17. Li Y, Wang C, Allen KE, et  al. Diagnosis of canine Hepatozoon 2007;93:283-299.
spp. infection by quantitative PCR. Vet Parasitol. 2008;157:50-58. 39. Baneth G, Shkap V. Monozoic cysts of Hepatozoon canis. J Parasi-
18. Gondim LF, Kohayagawa A, Alencar NX, et al. Canine hepatozo- tol. 2003;89:379-381.
onosis in Brazil: description of eight naturally occurring cases. Vet 40. Murata T, Makoto I, Susumu T, et  al. Vertical transmission of
Parasitol. 1998;74:319-323. Hepatozoon canis in dogs. J Vet Med Sci. 1993;55:867-868.
19. Eiras DF, Basabe J, Scodellaro CF, et  al. First molecular charac- 41. Johnson EM, Allen KE, Panciera RJ, et  al. Infectivity of Hep-
terization of canine hepatozoonosis in Argentina: evaluation of atozoon americanum cystozoites for a dog. Vet Parasitol.
asymptomatic Hepatozoon canis infection in dogs from Buenos 2008;154:148-150.
Aires. Vet Parasitol. 2007;149:275-279. 42. Johnson EM, Allen KE, Panciera RJ, et al. Experimental transmis-
20. Criado-Fornelio A, Rey-Valeiron C, Buling A, et al. New advances sion of Hepatozoon americanum to New Zealand White rabbits
in molecular epizootiology of canine hematic protozoa from Ven- (Oryctolagus cuniculus) and infectivity of cystozoites for a dog. Vet
ezuela, Thailand, and Spain. Vet Parasitol. 2007;144:261-269. Parasitol. 2009;164:162-166.
21. Gossett KA, Gaunt SD, Aja DS. Hepatozoonosis and ehrlichiosis in 43. Johnson EM, Panciera RJ, Allen KE, et  al. Alternate pathway of
a dog. J Am Anim Hosp Assoc. 1985;21:265-267. infection with Hepatozoon americanum and the epidemiologic
22. Baker JL, Craig TM, Barton CC, et al. Hepatozoon canis in a dog importance of predation. J Vet Intern Med. 2009;23:1315-1318.
with oral pyogranulomas and neurologic disease. Cornell Veteri- 44. Ewing SA, Mathew JS, Panciera RJ. Transmission of Hepatozoon
narian. 1988;78:179-183. americanum (Apicomplexa: Adeleorina) by ixodids (Acari: Ixodi-
23. Macintire DK, Vincent-Johnson N, Dillon AR, et al. Hepatozoonosis in dae). J Med Entomol. 2002;39(4):631-634.
dogs: 22 cases. J Am Vet Med Assoc. 1989-1994;1997(210):916-922. 45. Allen K, Yabsley M, Johnson E, et  al. Novel Hepatozoon
24. Panciera RJ, Gatto NT, Crystal MA, et al. Canine hepatozoonosis in vertebrates from the southern United States. J Parasitol.
in Oklahoma. J Am Anim Hosp Assoc. 1997;33:221-225. 2011;97:648-653.
25. Macintire DK, Vincent-Johnson NA. Canine hepatozoonosis. In: 46. Davis DS, Robinson RM, Craig TM. Naturally occurring hepato-
Bonagura, ed. Kirk’s Current Veterinary Therapy XIII Small Animal zoonosis in a coyote. J Wildl Dis. 1978;14:244-246.
Practice. Philadelphia, PA: WB Saunders; 2000:310-313. 47. Lane JR, Kocan AA. Hepatozoon sp. infection in bobcats. J Am Vet
26. Vincent-Johnson NA, Macintire DK, Lindsay DS, et  al. A new Med Assoc. 1983;183:1323-1324.
Hepatozoon species from dogs: description of the causative 48. Mercer SH, Jones LP, Rappole JH, et al. Hepatozoon sp. in wild
agent of canine hepatozoonosis in North America. J Parasitol. carnivores in Texas. J Wildl Dis. 1988;24:574-576.
1997;83(6):1165-1172. 49. Kocan AA, Breshears M, Cummings C, et  al. Naturally occur-
27. Baneth G, Barta JR, Shkap V, et  al. Genetic and antigenic evi- ring hepatozoonosis in coyotes from Oklahoma. J Wildl Dis.
dence supports the separation of Hepatozoon canis and Hepato- 1999;35(1):86-89.
zoon americanum at the species level. J Clin Microbiol. 2000;3: 50. Kocan AA, Cummings CA, Panciera RJ, et al. Naturally occurring
1298-1301. and experimentally transmitted Hepatozoon americanum in coy-
28. Dear JD, Owens SD, Biondo A, et al. Babesia conradae infection in otes from Oklahoma. J Wildl Dis. 2000;36(1):149-153.
coyote hunting dogs infected with multiple blood-borne pathogens. 51. Panciera RJ, Ewing SA, Mathew JS, et  al. Canine hepatozoono-
Proceedings of the 2012 American College of Veterinary Internal sis: comparison of lesions and parasites in skeletal muscle of dogs
Medicine Forum. New Orleans: LA; June 2012. experimentally or naturally infected with Hepatozoon america-
29. Little L, Baneth G. Cutaneous Hepatozoon canis infection in a dog num. Vet Parasitol. 1999;82:261-272.
from New Jersey. J Vet Diagn Invest. 2011;23:585-588. 52. Baneth G, Weigler B. Retrospective case-control study of hepatozo-
30. Baneth G, Macintire DK, Vincent-Johnson NA, et al. Hepatozoono- onosis in dogs in Israel. J Vet Intern Med. 1997;11:365-370.
sis. In: Greene, ed. Infectious Diseases of the Dog and Cat, 3rd ed. 53. Barton CL, Russo EA, Craig TM, et al. Canine hepatozoonosis: a
Philadelphia, PA: WB Saunders; 2006:698-711. retrospective study of 15 natural occurring cases. J Am Anim Hosp
31. Baneth G, Sheiner A, Eyal O, et al. Hepatozoon felis infection in Assoc. 1985;21:125-134.
domestic cats – morphologic description and genetic characteriza- 54. Gavazza A, Bizzeti M, Papini R. Observations on dogs found nat-
tion. Proceedings of the 2nd Symposium of the International Soci- urally infected with Hepatozoon canis in Italy. Revue Med Vet.
ety for Companion Animal Infectious Diseases. San Francisco: CA; 2003;154:565-571.
2012; Abstract O16. 55. Drost WT, Cummings CA, Mathew JS, et  al. Determination of
32. Mathew JS, Ewing SA, Panciera RJ, et al. Experimental transmis- time of onset and location of early skeletal lesions in young dogs
sion of Hepatozoon americanum to dogs by the Gulf Coast tick, experimentally infected with Hepatozoon americanum using bone
Amblyomma maculatum Koch. J Parasitol. 1998;80:1-14. scintigraphy. Vet Radiol Ultrasound. 2003;44:86-91.
33. Baneth G, Samish M, Alekseev E, et  al. Transmission of Hepa- 56. Panciera RJ, Mathew JS, Ewing SA, et al. Skeletal lesions of canine
tozoon canis to dogs by naturally fed or percutaneously injected hepatozoonosis caused by Hepatozoon americanum. Vet Pathol.
Rhipicephalus sanguineus ticks. J Parasitol. 2001;87:606-611. 2000;37:225-230.
CHAPTER 77  Canine and Feline Hepatozoonosis 759

57. Baneth G. Perspectives on canine and feline hepatozoonosis. Vet 66. Macintire DK, Vincent-Johnson NA, Kane CW, et  al. Treatment
Parasitol. 2011;181:3-11. of dogs infected with Hepatozoon americanum: 53 cases (1989-
58. Mathew JS, Saliki JT, Ewing SA, et al. An indirect enzyme-linked 1998). J Am Vet Med Assoc. 2001;218:77-82.
immunosorbent assay for diagnosis of American canine hepatozo- 67. Companion Animal Parasite Council Current Advice on Para-
onosis. J Vet Diagn Invest. 2001;13:17-21. site Control. Vector-borne Diseases: American Hepatozoonosis.
59. Shkap V, Baneth G, Pipano E. Circulating antibodies to Hepa- 2012. http://www.capcvet.org/capc-recommendations/american-
tozoon canis demonstrated by immunofluorescence. J Vet Diagn hepatozoonosis/. Last accessed January 21, 2013.
Invest. 1994;6:121-123. 68. Sasanelli M, Paradies P, Greco B, et al. Failure of imidocarb dipro-
60. Baneth G, Shkap V, Presentey BZ, et  al. Hepatozoon canis: the pionate to eliminate Hepatozoon canis in naturally infected dogs
prevalence of antibodies and gamonts in dogs in Israel. Vet Res based on parasitological and molecular evaluation methods. Vet
Commun. 1996;20:41-46. Parasitol. 2010;171:194-199.
61. Gonen L, Strauss-Ayali D, Shkap V, et al. An enzyme-linked immu- 69. Pasa S, Voyvoda H, Karagenc T, et al. Failure of combination therapy
nosorbent assay for antibodies to Hepatozoon canis. Vet Parasitol. with imidocarb dipropionate and toltrazuril to clear Hepatozoon canis
2004;122:131-139. infection in dogs. Parasitol Res. 2011;109:919-926.
62. Mylonakis ME, Leontides L, Gonen L, et  al. Anti-Hepatozoon 70. Carlos ET, Cruz FB, Cabiles CC, et al. Hepatozoon sp. in the WBC
canis serum antibodies and gamonts in naturally-occurring canine of a human patient. Univ Philipp Vet. 1971;15:5-7.
monocytic ehrlichiosis. Vet Parasitol. 2005;129:229-233. 71. Ewing GO. Granulomatous cholangiohepatitis in a cat due to
63. Auburn University College of Veterinary Medicine Molecular Diag- a protozoan parasite resembling Hepatozoon canis. Feline Pract.
nostics. http://www.vetmed.auburn.edu/molecular-diagnostics. Last 1977;7:37-40.
accessed January 21, 2013. 72. Baneth G, Lavy E, Presentey BZ. Hepatozoon sp. parasitemia in a
64. Vincent-Johnson NA. American canine hepatozoonosis. Vet Clin domestic cat. Feline Pract. 1995;23:10-12.
North Am Small Anim Pract. 2003;33:905-920.
65. Baneth G, Vincent-Johnson N. Hepatozoonosis. In: Shaw SE, Day
MJ, eds. Arthropod-borne Infectious Diseases of the Dog and Cat.
London: Manson; 2005:78-88.
CHAPTER 78

Trypanosomiasis
Stephen C. Barr, Ashley B. Saunders, and Jane E. Sykes

neurologic disease.2-7 However, many infected dogs are sub-


Overview of American Trypanosomiasis clinically infected for life. Cats can become infected, but dis-
First Described: Chagas’ disease (American trypanosomiasis) ease in cats has not been recognized. Trypanosoma cruzi is an
was first described in humans in 1909 (Carlos Chagas).1 important vector-borne zoonosis in the Americas, particularly
South and parts of Central America, and is the leading cause of
Cause: Trypanosoma cruzi, a protozoan parasite (phylum
dilated cardiomyopathy in humans.8 Within the United States,
Sarcomastigophora, family Trypanosomatidae)
most cases in dogs occur in Texas and especially involve work-
Affected Hosts: Dogs, humans, and more than 150 other ing dogs that reside in southeastern Texas (Figure 78-1).9-11
domestic or wildlife mammalian species A few affected dogs have been reported from other southern
Geographic Distribution: Central and South America; to a states,5,6,12-14 and as far north as Missouri.15 The seroprevalence
lesser extent southern United States (especially south- in dogs from Louisiana and Texas has generally been in the
eastern Texas) range of 12% to 22%, with seroprevalences of <10% in other
states where canine disease has been identified.16 Canine Cha-
Mode of Transmission: A bite from, or ingestion of, reduviid
gas’ disease is of importance to veterinary practitioners because
(kissing bug) vectors, primarily Triatoma species. Trans-
it can be difficult to diagnose, is a potential zoonosis, and there
mission through blood transfusion and vertical transmis-
is a lack of therapeutic options.
sion can occur.
Trypanosoma cruzi exists in three morphologic forms. The
Major Clinical Signs: American trypanosomiasis in pup- form that circulates freely in the host’s peripheral blood is the
pies manifests as lethargy, inappetence, mucosal pallor, trypomastigote. This form is 15 to 20 µm long, with a flattened
lymphadenopathy, splenomegaly, arrhythmias, or sud- spindle-shaped body and a centrally placed vesicular nucleus.
den death. Neurologic signs such as pelvic limb ataxia A single flagellum originates near a large, subterminal kineto-
and exaggerated spinal reflexes may also occur. Adult plast (situated posterior to the nucleus) and passes along the
dogs may show lethargy, arrhythmias, signs of right- or body to project anteriorly (Figure 78-2). The host intracellular
left-sided congestive heart failure, or sudden death. or amastigote form is approximately 1.5 to 4.0 µm in diameter,
Differential Diagnoses: The differential diagnoses for sus- is roughly spheroid, and contains both a nucleus and a rod-like
pected Chagas’ myocarditis include dilated cardiomy- kinetoplast. A small flagellum is present, but this is rarely obvi-
opathy of large-breed dogs, parvoviral myocarditis in ous under light microscopy. The third morphologic form, the
puppies, canine monocytic ehrlichiosis, babesiosis, and epimastigote, is found in the arthropod vector, a reduviid or
leishmaniosis. The primary differential diagnoses for “kissing bug” (subfamily Triatomae). Epimastigotes are flagel-
suspected Chagas’ meningoencephalitis in puppies are lated and spindle shaped with the kinetoplast situated anterior to
canine distemper virus infection and neosporosis, but the nucleus. When a vector is involved in transmission, infection
other infectious and inflammatory causes of meningoen- occurs when trypomastigotes are deposited in the triatomine’s
cephalitis should also be considered. feces at the insect bite site. This is the main mode of transmission
to humans in South America.
Human Health Significance: Transmission to humans has the Transmission of T. cruzi in endemic countries depends
potential to occur after needle-stick injuries; caution is on the confluence of vectors, reservoirs, parasites, and hosts
advised when handling tissues or clinical specimens from (both people and animals) in a single habitat. Only three Tri-
dogs with suspected trypanosomiasis. atomae species (Triatoma infestans [the main vector in South
America], Triatoma dimidiata, and Rhodnius prolixus) that
feed on man in endemic regions in South America display the
appropriate behavior that enables them to transmit T. cruzi
AMERICAN TRYPANOSOMIASIS effectively. These species feed on blood from both people and
domestic reservoir mammals (dog, cat, guinea pigs), reproduce
prolifically while cohabiting close to people, and defecate soon
Etiology and Epidemiology after taking a blood meal, meaning that they are usually still
Chagas’ disease, or American trypanosomiasis, is caused by the on the host near the bite wound when they defecate.17 Infec-
flagellated protozoan Trypanosoma cruzi. In North America, tion rates in these vectors can be as high as 100% south of the
disease in dogs usually manifests as cardiac disease typified equator. By contrast, domestic transmission cycles probably do
by arrhythmias or myocarditis (acute or chronic), and, rarely, not occur in the United States, except in areas of southeastern

760
CHAPTER 78  Trypanosomiasis 761

FIGURE 78-2  Trypomastigotes of Trypanosoma cruzi in a blood smear of a dog.


Wright-Giemsa stain, 1000× oil magnification. (From Barr SC. Canine Chagas’ disease
(American trypanosomiasis) in North America. Vet Clin North Am. 2009;39:1055-1064.)

or drinks.22 Blood transfusion and transplacental transmission


can also occur, and infection after ingestion of infected milk
from lactating bitches has been proposed.6 Whether ingestion
of reservoir hosts leads to transmission is unclear.
Within the United States, the principal wildlife reservoir
hosts of T. cruzi in the eastern seaboard states south from
A Maryland and in most other southern states (Texas, Louisiana,
and Oklahoma, to name a few) are opossums and raccoons.
Armadillos can also be infected.23 Various mouse, squirrel, and
rat species are the main reservoir hosts in New Mexico and Cal-
ifornia.24 Isolates of T. cruzi from vectors and animal reservoirs
in North America are less pathogenic in mice than South Ameri-
can isolates.23,25 Experimental inoculation of T. cruzi isolates
from opossums and armadillos into dogs produces a disease
consistent with naturally acquired acute and chronic canine try-
panosomiasis, so it is likely that dogs in nature are infected with
the same isolates as these wildlife hosts.2-6 Six genotypes of T.
cruzi have been identified (TcI through TcVI, also known as
discrete typing units or DTUs), which appear to differ in their
vector preferences and the extent to which they cause disease.26
After infection, trypomastigotes enter macrophages, transform
into amastigotes, and multiply by binary fission (Figure 78-3).
B Alternatively, they spread hematogenously from the site of
FIGURE 78-1  A, Approximate distribution of Chagas’ disease in humans in the infection, infect myocardiocytes, transform into amastigotes,
Americas. Insect-transmitted human Chagas’ disease has also been rarely reported in Ten- multiply, and transform back into trypomastigotes. The host
nessee, California and Louisiana. B, States within the United States where Chagas’ disease cell then ruptures and trypomastigotes are released back into
is most prevalent in dogs. Disease in Texas primarily occurs in the southeast. Occasional circulation. Parasitemia in dogs appears as early as 3 days
cases have been reported in other southern states and as far north as Virginia and Missouri. postinfection (DPI), peaks at 17 DPI, and is cytologically
undetectable (subpatent infection) by 30 DPI.2 Clinical signs
Texas where there is evidence to suggest that the dog can be of acute myocarditis, should they occur, develop about 14 DPI
involved in domestic transmission cycles involving vectors and with recovery occurring around 28 DPI.2 Rapid intracellular
man.18 Generally, though, the two principal vectors in the multiplication ensures a rapid rise in parasitemia before effec-
United States (Triatoma protracta and Triatoma sanguisuga) tive immunity develops. Parasitemia steadily rises as more and
have low infection rates (20%), display different feeding hab- more intracellular multiplication cycles add to the number
its, and defecate about 20 minutes after feeding, often when of circulating trypomastigotes. The vector becomes infected
they have long fled the host.19 As a result, it is more likely that when it ingests circulating trypomastigotes, which transform
dogs in North America become infected when they eat infected into epimastigotes and multiply by binary fission. Transforma-
triatomines or their excreta. Certainly, opossums20 and arma- tion of the epimastigotes back into trypomastigotes occurs in
dillos21 become infected by this route, and outbreaks have the vector’s hindgut before the trypomastigotes are passed in
occurred in humans following ingestion of contaminated food the feces.
762 SECTION 4  Protozoal Diseases

abnormalities become more prevalent in these dogs and may


even result in sudden death. Clinical signs referable to right-sided
and eventually, in some, left-sided heart failure occur and can
include exercise intolerance, pulse deficits, ascites, pleural effu-
sion, hepatomegaly, and jugular venous congestion.2 Chronic
Chagas’ myocarditis is clinically indistinguishable from dilated
cardiomyopathy of large breed dogs.4-6 The pathogenesis of the
cardiomyopathy is unknown, but proposed mechanisms include
damage to myocardiocytes or the autonomic nervous system by
immune-mediated mechanisms or toxic parasitic products, and/or
microvascular disease coupled with platelet dysfunction.27,28
Cardiac dilatation occurs when fibrosis no longer permits effi-
cient compensatory hypertrophy.27,29 Some T. cruzi isolates that
infect dogs in the United States are not pathogenic but produce
a marked serologic response and a low-level parasitemia fol-
lowing immunosuppression.2,30 Reactivation of infection with
immunosuppression can occur in human patients.22

Physical Examination Findings


The most common physical examination findings in puppies with
Chagas’ disease are lethargy, generalized lymphadenomegaly,
splenomegaly, pallor, and arrhythmias. Generalized subcutaneous
edema has also been reported.31 Dogs with chronic Chagas’ disease
may show signs of right- and/or left-sided congestive heart failure,
with tachypnea, ascites, or jugular pulses. Cardiac murmurs and/
or arrhythmias may be detected on auscultation of the heart.
FIGURE 78-3  Life cycle of Trypanosoma cruzi. Dogs are infected when a triatome
(reduviid bug) defecates at its bite site, and possibly when dogs ingest infected triatomes.
Other routes of transmission may also predominate in the United States (see text). Trypo-
Diagnosis
mastigotes infect cardiomyocytes (as well as other tissues) and form amastigotes, which
replicate locally, leading to myocarditis. The key to diagnosis of Chagas’ disease is a high degree of clini-
cal suspicion. Chagas’ disease should be considered in any dog
with signs of myocarditis or cardiomyopathy, particularly if it
Clinical Features lives or has lived at any time—even years before evaluation—in
an endemic region.
Signs and Their Pathogenesis
As in humans, there are three phases of Chagas’ myocarditis in Laboratory Abnormalities
dogs: acute, indeterminate (or latent), and chronic.2-6 Acute myo- Complete Blood Count
carditis results from cell damage and inflammation as trypomasti- During the acute T. cruzi infection, the CBC of some dogs may
gotes rupture from myocardiocytes. Lethargy, fever, inappetence, reveal anemia, leukocytosis due to a neutrophilia, eosinophilia,
generalized lymphadenopathy, slow capillary refill time with monocytosis, and lymphocytosis.32,33 Thrombocytopenia, leu-
pale mucus membranes, and, in some cases, splenomegaly and kopenia, and/or lymphopenia have also been documented.16
hepatomegaly are the main signs in puppies. Diarrhea can also Dogs in the chronic phase of infection may have no hematologic
occur. In dogs over 6 months of age, parasitemia develops more abnormalities.
slowly and clinical signs are often much less severe or not appar-
ent at all. Sudden death, presumably from cardiac muscle failure Serum Biochemical Tests
or conduction system failures leading to malignant arrhythmias, Serum ALT activity, AST activity, and creatinine and urea
is not a common occurrence. Although less common than signs nitrogen concentrations can be elevated in dogs with T. cruzi
referable to cardiac abnormalities, neurologic signs referable to infection, especially those that are at risk of death from severe
meningoencephalitis (as a direct result of parasitic invasion of the acute myocarditis. Rarely, moderate to severe hypoalbuminemia
neurologic system) may also occur and include weakness, pelvic with or without hyperglobulinemia occurs in acutely affected
limb ataxia, and exaggerated spinal reflexes suggestive of distem- dogs.16,31 Serum troponin I levels rise slowly in infected dogs
per. Humans with Chagas’ disease can also develop severe mega- and peak at around 10 to 30 mg/mL by 21 DPI. Serum troponin
esophagus or megacolon due to parasympathetic denervation,22 I levels are elevated in dogs infected after 6 months of age but
but these have not been described in dogs. usually not to such high levels.
Dogs that survive the acute phase enter a prolonged inde-
terminate phase typified by a lack of clinical signs. Parasitemia Urinalysis
becomes cytologically undetectable at about 30 DPI, although There are no specific findings reported on urinalysis in dogs
it can still be demonstrated by blood culture. The ECG is usu- with Chagas’ disease.
ally normal during this phase, although ventricular arrhythmias
can be induced by exercise.3 Although not all dogs progress to Electrocardiography
develop chronic disease, some develop chronic myocarditis with The ECG of dogs with severe Chagas’ myocarditis may show sinus
cardiac dilatation over the subsequent 8 to 36 months.2,3 ECG tachycardia, decreased R-wave amplitude, axis shifts, T-wave
CHAPTER 78  Trypanosomiasis 763

B
FIGURE 78-4  Electrocardiographic findings in dogs with Chagas’ disease. A, Second-degree heart block and depressed QRS complexes. (From Barr SC. Canine Chagas’ disease (Ameri-
can trypanosomiasis) in North America. Vet Clin North Am. 2009;39:1055-1064.)B, Multiform ventricular premature complexes in an 11-year-old, neutered male Labrador retriever present-
ing for collapse. Recording speed 25 mm/s, sensitivity 10 mm/mV.

inversion, ventricular arrhythmias, and conduction abnormali- effusions. Amastigotes were described in the lymph node of
ties, including first-, second-, and third-degree atrioventricular one affected dog that were cytologically indistinguishable from
block and right bundle branch block (Figure 78-4). those of Leishmania.31

Diagnostic Imaging Culture


Plain Radiography Trypanosoma cruzi can be cultured from blood in liver infu-
Plain thoracic radiographs in dogs with chronic Chagas’ myo- sion tryptose (LIT) growth medium. However, several weeks are
carditis may reveal cardiomegaly or evidence of congestive heart required before epimastigotes grow in this medium, which is too
failure (pulmonary edema or pleural effusion) (Figure 78-5). long to be helpful for making treatment decisions. Although cul-
ture is more sensitive than blood smear examination, false-neg-
Sonographic Findings ative results can still occur; in humans the sensitivity of blood
Echocardiograms of puppies with acute myocarditis are usu- culture is estimated to be only 50%.22 Culture of trypanosomes
ally within normal limits. Echocardiographic abnormalities in is a specialized procedure that is not routinely available in vet-
dogs with chronic Chagas’ myocarditis include right ventricular erinary diagnostic laboratories.
dilation with progression to include a loss of left ventricular
function with decreased fractional shortening, reduced ejec- Serologic Diagnosis
tion fraction, reduced left ventricular free wall thickness, and Serology is extremely useful for the diagnosis of Chagas’ dis-
increased end-systolic volume (Figure 78-6). ease, especially during the indeterminate and chronic phases
when trypomastigotes are very difficult to detect.34 Indirect fluo-
Microbiologic Tests rescent antibody, ELISA, and radioimmunoprecipitation assays
Diagnostic assays for trypanosomiasis in dogs and cats are are most commonly used. A rapid immunochromatographic
described in Table 78-1. dipstick test has also been developed.35 Serologic tests confirm
the presence of antibodies to T. cruzi, but most cross-react with
Cytologic Examination antibodies to Leishmania. Further, in rare situations in dogs,
During acute disease, trypomastigotes may be detected on blood the clinical signs of Chagas’ disease and leishmaniasis overlap
smear examination (see Figure 78-2). However, only a few par- sufficiently that it is necessary to go to considerable lengths to
asites may be present on the entire slide, demanding diligent establish a diagnosis.31 Therefore a detailed history of the likeli-
examination, or some form of concentration technique may be hood of exposure to Leishmania must be known in order to
used. High-power (400×) examination of the buffy-coat layer accurately interpret serologic results. A higher antibody titer to
from a centrifuged microhematocrit tube may reveal the motile Trypanosoma than to Leishmania may support a diagnosis of
parasites. Examination of a thick-film buffy-coat smear stained trypanosomiasis. Positive serology in association with consistent
with either Wright or Giemsa stains is more sensitive than clinical signs is the most common means of diagnosis of Chagas’
examination of a blood smear. A highly effective concentra- disease in dogs. If possible, two different serologic assays should
tion technique involves pelleting trypomastigotes from plasma be used to confirm a positive titer, especially in regions of low
(obtained by centrifugation of 10 to 50 mL of heparinized blood prevalence. The serum titer usually becomes positive by 21 DPI,
at 800 × g for 10 minutes) by further centrifugation (8000 × g when parasitemia is declining, and persists for the life of the
for 15 minutes). The pellet from the final centrifugation may animal irrespective of whether clinical signs develop.30
be examined microscopically after staining or be submitted for
PCR analysis or culture. The larger the volume of the blood Molecular Diagnosis Using the Polymerase Chain Reaction
specimen collected, the greater the likelihood that organisms PCR assays, which detect DNA of T. cruzi in various clinical
will be detected. Trypomastigotes may also be found on cyto- specimens (blood, plasma pellets, lymph node aspirates, or
logic examination of lymph node aspirates and in abdominal ascites fluid), can be highly specific for T. cruzi but can have
764 SECTION 4  Protozoal Diseases

A
A

B
FIGURE 78-5  Right lateral (A) and dorsoventral (B) thoracic radiographic images of
a dog with trypanosomiasis and mild generalized cardiomegaly. The pulmonary veins are
slightly larger than the corresponding artery, and a slight interstitial pattern was present
throughout the lungs.
C
variable and sometimes low sensitivity for diagnosis of chronic FIGURE 78-6  Two dimensional (A) and M-mode (B) transthoracic echocardiogram
infections unless multiple specimens are tested.34 Real-time images obtained in a right parasternal short-axis view and long-axis view (C) from a dog
PCR assays have been developed for diagnosis of human Cha- with Chagas’ disease documenting significant left ventricular enlargement and mild right
ventricular enlargement.
gas’ disease that detect repetitive sequences found in very high
copy number within T. cruzi.36 Because each parasite may
have more than 100,000 copies of the sequence, the sensitivity Pathologic Findings
of the assay is increased.22 The sensitivity of PCR assays var- Gross Pathologic Findings
ies depending on the T. cruzi strain involved.36,37 PCR may Gross pathologic findings in dogs with Chagas’ disease include
be useful to confirm infection when organisms are not seen lymphadenopathy, evidence of congestive heart failure (ascites,
on blood smears. Currently, PCR assays for T. cruzi are not pleural effusion, or congestion of viscera), and biventricular car-
widely available from veterinary diagnostic laboratories on a diac enlargement with thinning of the ventricular wall. Often the
commercial basis, but have been used for diagnosis of canine right side of the heart is more enlarged than the left. Hemorrhages
Chagas’ disease.31 and pale foci may be identified in the myocardium.
CHAPTER 78  Trypanosomiasis 765

TABLE 78-1
Diagnostic Assays Currently Available for Trypanosomiasis in Dogs and Cats
Assay Specimen Type Target Performance
Cytologic examination Aspirates of lymph nodes, Trypanosoma Organisms may be few in number and in chronic
spleen, bone marrow, trypomastigotes or, trypanosomiasis are often not found.
blood smears, or buffy- less commonly,
coat preparations amastigotes
Trypanosoma culture Blood or buffy-coat Trypanosoma spp. Requires special techniques and expertise that are
preparations, bone not widely available. Sensitivity may be low in
marrow chronic infections.
Antibody serology Serum Antibodies to Most infected dogs test positive, but positive test
(immunofluorescent Trypanosoma spp. results occur in healthy animals, so results must
antibody or ELISA) be interpreted in light of clinical signs. False posi-
tives can occur in dogs exposed to Leishmania or
those vaccinated with Leishmania vaccines.
Real-time PCR assays Whole blood, buffy coats, Trypanosoma DNA May be useful in dogs with suggestive clinical signs
splenic or lymph node in which organisms cannot be found cytologi-
aspirates, CSF cally. Sensitivity and specificity depend on assay
design. Quantitative PCR may be useful for evalu-
ation of treatment efficacy. Limited availability.

TABLE 78-2
Antimicrobials Used to Treat Acute Chagas’ Disease in Dogs
Dose Interval Duration
Drug* (mg/kg) Route (hours) (months)
Benznidazole 5-10 PO 24 2
Nifurtimox 2-7 PO 6 3-5
Prednisone 0.5 PO 12 1

*Both benznidazole and nifurtimox are available from the Centers for
Disease Control and Prevention, Atlanta, GA.

Treatment and Prognosis


Antimicrobial Treatment
Treatment of dogs with naturally occurring acute Chagas’
myocarditis is poorly documented, since this phase is seldom
recognized. The use of nifurtimox (usually in association with
FIGURE 78-7  Pseudocyst of Trypanosoma cruzi within a myocardiocyte of an infected glucocorticoids)38 or benznidazole39 has been reported in the
dog. H&E stain, 1000× magnification. (From Barr SC. Canine Chagas’ disease (American dog (Table 78-2). However, the severe adverse effects of nifur-
trypanosomiasis) in North America. Vet Clin North Am. 2009;39:1055-1064.) timox preclude its use, so benznidazole is the drug of choice
(see Chapter 10). Benznidazole is available from the Centers
for Disease Control and Prevention, Atlanta, GA. After treat-
Histopathologic Findings ment with benznidazole, serum antibody titers usually remain
Histopathologic findings in Chagas’ disease include a severe elevated, although they are reported to decline in people.
diffuse granulomatous myocarditis, large numbers of parasitic Treatment of experimentally infected dogs with acute Chagas’
pseudocysts (intracellular collections of amastigotes), and mini- disease with benznidazole reduced or prevented subsequent
mal fibrosis (Figure 78-7). Immunohistochemistry does not dis- fibrosis and arrhythmias, even if parasites were not completely
tinguish Leishmania from Trypanosoma amastigotes, because of eliminated.40 Benznidazole-resistant strains of T. cruzi have
serologic cross-reactivity.31 Histopathology of the myocardium been described. Ketoconazole, gossypol, allopurinol, and vera-
in chronic Chagas’ myocarditis is characterized by multifocal pamil have shown promise in other species but are ineffective
lymphocytic infiltrates, perivasculitis, and marked fibrosis, and for the treatment of Chagas’ disease in dogs. Ravuconazole and
rare, if any, parasitic pseudocysts.4,27 posaconazole also reduce parasite load,41,42 but to date azole
766 SECTION 4  Protozoal Diseases

drugs have not been adopted for treatment of humans because and Louisiana). By far the largest number of people infected
they lack significant benefit over benznidazole or nifurtimox.22 with T. cruzi in the United States (estimated at approximately
However, the combination of the antiarrhythmic amiodarone 100,000 people) have emigrated from endemic regions such as
and an azole (itraconazole or posaconazole) has synergistic Mexico and Central America. Consequently, reports of cases
antiprotozoal activity against T. cruzi and shows promise as an associated with transmission by blood transfusion continue to
effective treatment for chronic Chagas’ disease in humans.43,44 rise.51,52 Imported cases in tourists who return to the United
Most dogs are diagnosed with Chagas’ disease during the States have not been described. There are probably several rea-
chronic stage. Unfortunately, treatments directed against the sons why only a handful of naturally acquired cases have been
parasite at this stage do not significantly alter the outcome of reported in the United States. First, North American vector spe-
disease.45 Treatment should be directed toward the myocar- cies are poorly adapted to living in houses and do not defecate
dial failure and ventricular arrhythmias, although the latter are on the host after a blood meal. Higher standards of housing in
often refractory to drug therapy. Pacemaker implantation may North America than in parts of Central and South America may
be required for some dogs with bradyarrhythmias such as third- also prevent vectors from nesting in human dwellings. Third,
degree atrioventricular block. In humans with indeterminate and some human cases of Chagas’ disease in North America may be
chronic symptomatic Chagas’ disease, the benefit of treatment overlooked because of a low level of suspicion.
remains controversial and is still under investigation by means Although the risk of a human acquiring infection from an
of a large, prospective clinical trial (the Benznidazole Evaluation infected dog is extremely low, the severity and difficulty of
for Interrupting Trypanosomiasis [BENEFIT] trial).36 treating disease in humans makes this disease of considerable
public health significance. Veterinarians should be careful when
Prognosis handling blood samples from infected dogs and warn labora-
The prognosis for dogs that develop Chagas’ disease is poor tory staff of the potential infectivity of the samples. Accidental
because of the lack of effective treatments. Dogs diagnosed at needle-stick injuries when administering therapy to or collecting
an older age (mean of 9 years) survive longer (30 to 60 months) specimens from infected dogs should be reported immediately
than dogs diagnosed at a younger age (mean of 4.5 years), which to the CDC.
survive only up to 5 months postdiagnosis.11

Immunity and Vaccination Overview of African Trypanosomiasis in Dogs


First Described: Reports of African trypanosomiasis date back
Recovery from T. cruzi infection is not associated with protective
to Egyptian times. Trypanosomes were identified as the
immunity. No vaccine is available.
cause in 1895, when they were detected in cattle by a
Scottish microbiologist, David Bruce (after whom Brucella
Prevention was named).53
The risk of infection by T. cruzi can be reduced by limiting con- Cause: Trypanosoma brucei rhodesiense, Trypanosoma con-
tact between dogs and vectors and possible reservoir hosts (rac- golense, Trypanosoma evansi (protozoan parasites, family
coons, opossums, armadillos, and skunks). Dogs should not be Trypanosomatidae)
fed meat from reservoir hosts. Kennels and surrounding struc- Affected Hosts: Dogs and a variety of other mammalian host
tures (chicken houses, wood piles) in endemic areas should be species. Humans are infected with T. brucei gambiense and
sprayed monthly with a residual insecticide. Dog housing should T. brucei rhodesiense.
be upgraded to remove vector nesting sites. Applications of
fipronil on the coats of dogs do not appear to prevent infections Geographic Distribution: Central Africa (T. brucei, T. congolense);
in dogs or reduce the feeding of vectors,46 but deltamethrin- T. evansi is found in Africa as well as in Asia, Latin America,
treated collars do reduce feeding by Triatoma infestans.47,48 and the Canary Islands in Spain.
Blood donor dogs should be serologically screened to determine Mode of Transmission: A bite from the tsetse fly (T. brucei or
previous exposure to T. cruzi. In highly endemic regions (south- T. congolense) or mechanical transmission by hematopha-
ern Texas), bitches should be screened serologically, and posi- gous flies (T. evansi)
tive animals should not be bred. Major Clinical Signs: Lethargy, inappetence, weight loss, pal-
lor, peripheral edema, ascites, hematemesis, hemorrhagic
Public Health Aspects diarrhea, uveitis, corneal edema, neurologic signs due to
meningoencephalitis
American trypanosomiasis is a major human health problem in
South and Central America and is increasingly recognized in Differential Diagnoses: These include canine monocytic
Mexico.8,49 It is primarily a problem in impoverished regions ehrlichiosis, babesiosis, leishmaniosis, and systemic
where humans live in rural areas. Dogs are considered a reser- immune-mediated diseases. Canine distemper virus
voir for human infection in these regions because they maintain infection and neosporosis are important differential diag-
high levels of parasitemia and are a preferred host for triatomine noses in dogs with meningoencephalitis.
insects. Cats may also act as a reservoir for human infection Human Health Significance: Although most Trypanosoma
in South America.50 Fewer than one third of infected humans species that cause African trypanosomiasis in dogs do
ultimately develop Chagas’ disease.22 not infect humans, T. brucei rhodesiense does, so caution is
To date, fewer than 10 human cases of Chagas’ disease advised when handling tissues or clinical specimens from
involving transmission by vectors have been reported in the dogs or cats with suspected trypanosomiasis.
United States (4 in Texas, and 1 each in California, Tennessee,
CHAPTER 78  Trypanosomiasis 767

AFRICAN TRYPANOSOMIASIS glands of the tsetse fly, and the organism is transmitted in saliva
when it feeds on the mammalian host. Transmission to the mam-
African trypanosomiasis is caused by Trypanosoma brucei and malian host results in hemolymphatic spread of the organism over
Trypanosoma congolense. These organisms are transmitted by weeks to months with signs that relate to proliferation of mono-
tsetse flies (Glossina species), which are only found in Africa. Try- nuclear phagocytes and increased vascular permeability. T. brucei
panosoma brucei includes Trypanosoma brucei brucei, T. brucei complex organisms are well known for their ability to evade the
gambiense, and T. brucei rhodesiense, which are indistinguish- host immune response through extensive variation of their outer
able morphologically and are referred to as “T. brucei complex” surface glycoproteins (known as variant antigen types, or VATs).
organisms. T. evansi also belongs to this group (see later). The The hemolymphatic phase may be followed by the development
epidemiology, host range, and clinical features of disease caused of meningoencephalitis in some animals. In humans, meningoen-
by these T. brucei complex organisms differ. Trypanosoma brucei cephalitis is most apparent with T. brucei gambiense infections
gambiense and T. brucei rhodesiense cause “sleeping sickness” in and results in tremors and progressive daytime somnolescence
humans from Africa (West African and East African trypanoso- (hence the term “sleeping sickness”). Pancarditis with arrhyth-
miasis, respectively) (Figure 78-8). A variety of animal reservoirs mias and congestive heart failure is a more typical outcome of T.
of T. brucei rhodesiense have been identified, but an animal res- brucei rhodesiense infection in humans.
ervoir for T. brucei gambiense has not.54 Along these lines, only The clinical signs of African trypanosomiasis in dogs include
naturally occurring infections with T. brucei rhodesiense , and persistent fever, lethargy, anorexia, weight loss, pallor, muco-
not T. brucei gambiense, have been reported in dogs.55 Trypano- purulent oculonasal discharge, lymphadenopathy, hepatospleno-
soma brucei brucei and T. congolense cause trypanosomiasis in a megaly, variable peripheral edema, abdominal distention due to
variety of domestic animal species (“nagana”) but do not infect ascites, petechial hemorrhages, signs of pancarditis, and ocular
humans. Dogs are especially susceptible to T. congolense infec- signs such as unilateral or bilateral uveitis, corneal edema, and/
tion. Four types of T. congolense have been identified—savan- or keratitis (which can mimic “blue eye” due to canine adeno-
nah, forest, Tsavo, and Kilifi—which may vary in pathogenicity. virus-1). Hemorrhagic vomiting and diarrhea can occur in dogs
There are several reports of travel-related infections with T. con- infected with T. congolense.58,60 Neurologic signs (seizures, trem-
golense in dogs,56-58 sometimes in association with a long period ors, opisthotonos, and hyperreflexia) due to meningoencephalitis
of latency.57 It has been suggested that African dogs may exhibit a have also been described in dogs infected with T. congolense.58
degree of “trypanotolerance,” whereas dogs imported into Africa Laboratory abnormalities in dogs with African trypanosomi-
may be more susceptible to infection and disease.59 asis have been most well described for T. congolense infections.
African trypanosomes exist only as a trypomastigote in the The most common findings are regenerative or nonregenerative
blood of the mammalian host; in contrast to Chagas’ disease, anemia, leukocytosis or leukopenia, and thrombocytopenia.57,58,60
there is no amastigote phase. Epimastigotes develop in the salivary Serum biochemistry findings include increased serum liver

FIGURE 78-8  Map showing geographic distribution of human African trypanosomiasis (green countries) and the number of cases reported in natives in the year 2009. Trypanosoma brucei
gambiense is found in western Africa and Trypanosoma brucei rhodesiense is found in eastern Africa. The line divides the geographic distribution of the two forms of disease. T. brucei brucei,
which infects dogs, occurs throughout both regions. The hashed lines indicate the countries in which tsetse flies are found. (Modified from Brun R and Blum J. Human African Trypanosomiasis.
Inf Dis Clin North Am. 2012;26:261-273.)
768 SECTION 4  Protozoal Diseases

enzyme activities, azotemia, hyperglobulinemia, and hypoalbu- of kinetoplast DNA, which stops it from developing within the
minemia,57,58 but one dog had no significant abnormal serum insect vector. Instead, the organism is transmitted mechanically
biochemistry findings.56 by biting flies. Ingestion of infected meat may also lead to infec-
The primary means of diagnosis is cytologic detection of the tion. Because populations of biting flies are greatest during the
organism in aspirates, body fluids, or blood smears. Identification rainy season, a seasonal incidence of disease has been observed
of the infecting species requires PCR and sequencing. The treat- in dogs.62 Travel-related disease has been described.63,64 The
ment of choice for dogs is diminazene aceturate, but this may not disease has an acute course; clinical signs and laboratory abnor-
always be effective or available. Treatment of the hemolymphatic malities resemble those of African trypanosomiasis, with leth-
disease in humans is with pentamidine isethionate (gambiense) or argy, fever, anorexia, weight loss, pallor, lymphadenopathy,
suramin (rhodesiense), whereas disease of the central nervous sys- vomiting, conjunctivitis, uveitis, corneal edema, and peripheral
tem is treated with the arsenical melarsoprol, but these drugs can edema, as well as leukopenia, thrombocytopenia, and anemia on
have significant adverse effects. Pentamidine was used with appar- the CBC.62-65 Hyperglobulinemia and proteinuria have also been
ent success to treat one dog with African trypanosomiasis.61 Pre- reported.64 Some dogs develop neurologic signs late in the course
vention is through the use of insect repellants and indoor housing. of disease due to nonsuppurative meningoencephalitis.64 Defini-
tive diagnosis of infection is based on cytologic examination
OTHER TRYPANOSOMA INFECTIONS of blood smears and/or PCR assay. Treatment is usually with
diminazene aceturate or suramin, although diminazene aceturate
Other Trypanosoma species that can infect dogs (and to a lesser is considered less effective against T. evansi than T. congolense
extent, cats) include Trypanosoma evansi and Trypanosoma infection.64 Untreated infection is usually fatal in dogs.
caninum. Trypanosoma caninum is a recently discovered Trypanosoma
T. evansi causes surra (= “rotten”) and is endemic in large species that has only been identified in dogs from South America,
parts of Asia, Africa, Latin America, and the Canary Islands many of which have been apparently healthy.66,67 The pathogenic-
of Spain. It causes disease in a variety of mammalian host spe- ity of this organism requires further study. Importantly, infection
cies that include horses, cattle, dogs, and cats, but not humans. with T. caninum leads to false-positive serologic test results for
T. evansi is very closely related to T. brucei and may have Leishmania, which can confound control methods that rely on cull-
derived from T. brucei as a result of partial or complete loss ing of dogs that are seropositive for Leishmania (see Chapter 74).

CASE EXAMPLE MCHC 35 g/dL (32-36 g/dL)


WBC 13,100 cells/µL (6000-17,000 cells/µL)
Neutrophils 8777 cells/µL (3000-11,500 cells/µL)
Signalment: “Duke,” a 5-year-old male beagle mix dog from Lymphocytes 2096 cells/µL (1000-4800 cells/µL)
San Antonio, TX Monocytes 1179 cells/µL (150-1250 cells/µL)
History: Duke’s owner reported a 7-day history of inappetence, Platelets 238,000 platelets/µL (200,000-500,000 platelets/µL)
lethargy, and decreased exercise tolerance. Two months Serum Chemistry Profile:
earlier, the dog had been diagnosed with dirofilariasis and Sodium 144 mmol/L (139-147 mmol/L)
was treated with melarsomine dihydrochloride using a split Potassium 3.9 mmol/L (3.3-4.6 mmol/L)
protocol. He had not traveled out of his local area. He was Chloride 118 mmol/L (107-118 mmol/L)
up to date on vaccinations, which included vaccines for Bicarbonate 22 mmol/L (20-28 mmol/L)
distemper, hepatitis, parvovirus, and rabies. Phosphorus 3.6 mg/dL (2.9-6.2 mg/dL)
Current medications: Monthly oral ivermectin/pyrantel Calcium 9.8 mg/dL (9.3-11.8 mg/dL)
heartworm preventative. BUN 19 mg/dL (5-29 mg/dL)
Physical Examination: Creatinine 1.27 mg/dL (0.3-2.0 mg/dL)
Body weight: 19.9 kg. Glucose 108 mg/dL (60-135 mg/dL)
General: Lethargic. T = 101.7°F (38.7°C), HR = 142 beats/min, Total protein 5.5 g/dL (5.7-7.8 g/dL)
RR = 48 breaths/min, mucous membranes pink, CRT = 1 s. Albumin 2.6 g/dL (2.4-3.6 g/dL)
Integument, Eyes, Ears, Nose, and Throat: Full, shiny Globulin 2.9 g/dL (1.7-3.8 g/dL)
haircoat. No evidence of ectoparasites was detected. No ALT 136 U/L (10-130 U/L)
other significant abnormalities were noted. ALP 103 U/L (21-147 U/L)
Musculoskeletal: Body condition score was 6/9. GGT 11 U/L (0-25 U/L)
Cardiovascular: An irregular rhythm and a grade 3/6 systolic Cholesterol 199 mg/dL (120-247 mg/dL)
left apical murmur were detected on thoracic auscultation. Total bilirubin 0.2 mg/dL (0-0.8 mg/dL)
Strong femoral pulses were palpated bilaterally. Magnesium 2.1 mg/dL (1.7-2.1 mg/dL)
Respiratory: Mild tachypnea was identified with normal breath Urinalysis: SGr 1.012; pH 7.5, trace protein (SSA), no bilirubin,
sounds and normal respiratory effort. no glucose, no WBC/HPF, 0-1 RBC/HPF.
Gastrointestinal, Genitourinary, and Lymph Nodes: No Heartworm Antigen Test: Negative.
abnormalities were detected. Cardiac Troponin I: 0.69 ng/mL (<0.2 ng/mL).
Laboratory Findings: Chagas Immunofluorescent Antibody Titer: Positive at
CBC: 1:160.
PCV 48.3% (31%-56%)
MCV 66.4 fL (60-77 fL)
CHAPTER 78  Trypanosomiasis 769

Imaging Findings: (1 mg/kg PO q12h). This was associated with improvement


Thoracic Radiographs: The cardiac silhouette was mildly, in clinical signs within 1 week of starting treatment.
generally enlarged. The pulmonary veins were mildly larger Comments: This dog was diagnosed with trypanosomiasis
than the corresponding artery. A mild pulmonary interstitial based on an index of suspicion in the presence of ventricular
pattern was present. enlargement and dysfunction in combination with multiform
Echocardiogram: The left ventricular internal dimensions ventricular arrhythmias. Infection by Leishmania could also
were enlarged, and systolic function was reduced (fractional have caused the positive serologic test result, but this was
shortening 16.1%, area shortening 26.9%; normal is >25% considered unlikely based on the geographic location and
and >50%, respectively). Moderate mitral regurgitation was clinical signs. PCR assays or Trypanosoma blood culture could
documented, and the left atrium was enlarged. The right have been performed to confirm the diagnosis, but these are
ventricle was mildly dilated. Pulmonic and aortic velocities not widely available and have low sensitivity when used in
were within normal limits. No heartworms were visualized. chronic infections. Antiprotozoal drugs were not used to
Additional Testing: Electrocardiogram: Sinus rhythm with treat this dog because they do not appear to alter outcome
frequent, multiform ventricular premature complexes. in chronic Chagas’ disease. The dog did very well clinically
Blood pressure: Systolic blood pressure was 115 mm Hg. while being managed with cardiac medications including
Diagnosis: American trypanosomiasis (Chagas’ disease). antiarrhythmics along with routine evaluation. After
Treatment: Medication to address cardiac enlargement, 2.5 years, ventricular arrhythmias increased in severity and
dysfunction, and arrhythmias included pimobendan (0.25 were more difficult to control with antiarrhythmic therapy.
mg/kg PO q12h), enalapril (0.5 mg/kg PO q12h), and sotalol He died suddenly at home 5 days after reevaluation.

SUGGESTED READINGS 11. Meurs KM, Anthony MA, Slater M, et al. Chronic Trypanosoma
cruzi infection in dogs: 11 cases (1987-1996). J Am Vet Med Assoc.
Museux K, Boulouha L, Majani S, et al. African Trypanosoma infection 1998;213:497-500.
in a dog in France. Vet Rec. 2011;168:590. 12. Bradley KK, Bergman DK, Woods JP, et al. Prevalence of American
Nabity MB, Barnhart K, Logan KS, et  al. An atypical case of Try- trypanosomiasis (Chagas disease) among dogs in Oklahoma. J Am
panosoma cruzi infection in a young English Mastiff. Vet Parasitol. Vet Med Assoc. 2000;217:1853-1857.
2006;140:356-361. 13. Snider TG, Yaeger RG, Dellucky J. Myocarditis caused by Try-
panosoma cruzi in a native Louisiana dog. J Am Vet Med Assoc.
REFERENCES 1980;177:247-249.
1. Chagas C. Nova tripanosomiase humana. Estudos sobre a morfo- 14. Tippit TS. Canine trypanosomiasis (Chagas’ disease). Southwest
logia e o ciclo evolutivo do Schizotrypanum cruzi n. gen., n. sp., Vet. 1978;2:97-104.
agente etiologico de nova entradade morbida do homen. Mem Inst 15. Cohn L. Unpublished observations, 2012.
Oswaldo Cruz. 1909;1:1-9. 16. Patel JM, Rosypal AC, Zimmerman KL, et  al. Isolation, mouse
2. Barr SC, Gossett KA, Klei TR. Clinical, clinicopathologic, and pathogenicity, and genotyping of Trypanosoma cruzi from an English
parasitologic observations of trypanosomiasis in dogs infected Cocker Spaniel from Virginia, USA. Vet Parasitol. 2012;187:394-398.
with North American Trypanosoma cruzi isolates. Am J Vet Res. 17. Carcavallo RU. The subfamily Triatominae (Hemoptera, Reduvi-
1991;52:954-960. idae): systematics and ecological factors. In: Brenner RR, Stoke A, eds.
3. Barr SC, Holmes RA, Klei TR. Electrocardiographic and echo- Chagas’ Disease Vectors. Boca Raton, FL: CRC Press; 1987:13-18.
cardiographic features of trypanosomiasis in dogs inoculated 18. Beard CB, Pye G, Steurer FJ, et  al. Chagas disease in a domestic
with North American Trypanosoma cruzi isolates. Am J Vet Res. transmission cycle, southern Texas. USA. Emerg Infect Dis. 2003;9:
1992;53:521-527. 103-105.
4. Barr SC, Schmidt SP, Brown CC, et al. Pathologic features of dogs 19. Yaeger RG. The present status of Chagas’ disease in the United
inoculated with North American Trypanosoma cruzi isolates. Am J States. Bull Tulane Univ Med Fac. 1961;21:9-13.
Vet Res. 1991;52:2033-2039. 20. Yaeger RG. Transmission of Trypanosoma cruzi infection to opos-
5. Barr SC, Simpson RM, Schmidt SP, et al. Chronic dilatative myo- sums via the oral route. J Parasitol. 1971;57:1375-1376.
carditis caused by Trypanosoma cruzi in two dogs. J Am Vet Med 21. Roellig DM, Ellis AE, Yabsley MJ. Oral transmission of Trypano-
Assoc. 1989;195:1237-1241. soma cruzi with opposing evidence for the theory of carnivory.
6. Barr SC, Van Beek O, Carlisle-Nowak MS, et  al. Trypanosoma J Parasitol. 2009;95:360-364.
cruzi infection in Walker hounds from Virginia. Am J Vet Res. 22. Kirchhoff LV. Trypanosoma species (American Trypanosomiasis,
1995;56:1037-1044. Chagas’ disease): biology of trypanosomes. In: Mandell GL,
7. Berger SL, Palmer RH, Hodges CC, et  al. Neurologic manifesta- Bennett JE, Dolin R, eds. Principles and Practice of Infectious
tions of trypanosomiasis in a dog. J Am Vet Med Assoc. 1991;198: Diseases. 7th ed. Philadelphia, PA: Elsevier; 2010:3481-3488.
132-134. 23. Barr SC, Brown CC, Dennis VA, et al. The lesions and prevalence
8. Espinosa R, Carrasco HA, Belandria F, et al. Life expectancy anal- of Trypanosoma cruzi in opossums and armadillos from southern
ysis in patients with Chagas’ disease: prognosis after one decade Louisiana. J Parasitol. 1991;77:624-627.
(1973-1983). Int J Cardiol. 1985;8:45-56. 24. Woody NC, Woody HB. American trypanosomiasis. I. Clinical and
9. Barr SC. Canine American trypanosomiasis. Compend Cont Educ epidemiologic background of Chagas’ disease in the United States.
Pract Vet. 1991;13:745-755. J Pediatr. 1961;58:568-580.
10. Kjos SA, Snowden KF, Craig TM, et  al. Distribution and char- 25. Barr SC, Dennis VA, Klei TR. Growth characteristics in axenic and cell
acterization of canine Chagas disease in Texas. Vet Parasitol. cultures, protein profiles, and zymodeme typing of three Trypanosoma
2008;152:249-256. cruzi isolates from Louisiana mammals. J Parasitol. 1990;76:631-638.
770 SECTION 4  Protozoal Diseases

26. Zingales B, Miles MA, Campbell DA, et al. The revised Trypano- 44. Benaim G, Sanders JM, Garcia-Marchan Y, et al. Amiodarone has
soma cruzi subspecific nomenclature: rationale, epidemiological intrinsic anti–Trypanosoma cruzi activity and acts synergistically
relevance and research applications. Infect Genet Evol. 2012;12: with posaconazole. J Med Chem. 2006;49:892-899.
240-253. 45. Santos FM, Lima WG, Gravel AS, et al. Cardiomyopathy progno-
27. Andrade ZA, Andrade SG, Correa R, et  al. Myocardial changes sis after benznidazole treatment in chronic canine Chagas’ disease.
in acute Trypanosoma cruzi infection. Ultrastructural evidence of J Antimicrob Chemother. 2012;67:1987-1995.
immune damage and the role of microangiopathy. Am J Pathol. 46. Gurtler RE, Ceballos LA, Stariolo R, et al. Effects of topical appli-
1994;144:1403-1411. cation of fipronil spot-on on dogs against the Chagas disease vector
28. Andrade ZA, Andrade SG, Sadigursky M, et al. The indeterminate Triatoma infestans. Trans R Soc Trop Med Hyg. 2009;103:98-304.
phase of Chagas’ disease: ultrastructural characterization of car- 47. Reithinger R, Ceballos L, Stariolo R, et al. Chagas disease control:
diac changes in the canine model. Am J Trop Med Hyg. 1997;57: deltamethrin-treated collars reduce Triatoma infestans feeding suc-
328-336. cess on dogs. Trans R Soc Trop Med Hyg. 2005;99:502-508.
29. Tanowitz HB, Kirchhoff LV, Simon D, et al. Chagas’ disease. Clin 48. Reithinger R, Ceballos L, Stariolo R, et  al. Extinction of experi-
Microbiol Rev. 1992;5:400-419. mental Triatoma infestans populations following continuous expo-
30. Barr SC, Dennis VA, Klei TR, et  al. Antibody and lymphoblas- sure to dogs wearing deltamethrin-treated collars. Am J Trop Med
togenic responses of dogs experimentally infected with Trypano- Hyg. 2006;74:766-771.
soma cruzi isolates from North American mammals. Vet Immunol 49. Estrada-Franco JG, Bhatia V, Diaz-Albiter H, et  al. Human Try-
Immunopathol. 1991;29:267-283. panosoma cruzi infection and seropositivity in dogs, Mexico.
31. Nabity MB, Barnhart K, Logan KS, et al. An atypical case of Try- Emerg Infect Dis. 2006;12:624-630.
panosoma cruzi infection in a young English Mastiff. Vet Parasitol. 50. Cardinal MV, Lauricella MA, Ceballos LA, et al. Molecular epide-
2006;140:356-361. miology of domestic and sylvatic Trypanosoma cruzi infection in
32. Guedes PM, Veloso VM, Mineo TW, et al. Hematological altera- rural northwestern Argentina. Int J Parasitol. 2008;38:1533-1543.
tions during experimental canine infection by Trypanosoma cruzi. 51. Kirchhoff LV. American trypanosomiasis (Chagas’ disease)—a tropi-
Rev Bras Parasitol Vet. 2012;21:151-156. cal disease now in the United States. N Engl J Med. 1993;329:639-644.
33. Quijano-Hernandez IA, Castro-Barcena A, Aparicio-Burgos E, 52. Schmunis GA. Trypanosoma cruzi, the etiologic agent of Chagas’
et  al. Evaluation of clinical and immunopathological features of disease: status in the blood supply in endemic and nonendemic
different infective doses of Trypanosoma cruzi in dogs during the countries. Transfusion. 1991;31:547-557.
acute phase. Scientific World Journal. 2012;2012:635169. 53. Steverding D. The history of African trypanosomiasis. Parasit
34. Araujo FM, Bahia MT, Magalhaes NM, et al. Follow-up of experi- Vectors. 2008;1:3.
mental chronic Chagas’ disease in dogs: use of polymerase chain 54. Balyeidhusa AS, Kironde FA, Enyaru JC. Apparent lack of a domes-
reaction (PCR) compared with parasitological and serological tic animal reservoir in Gambiense sleeping sickness in northwest
methods. Acta Trop. 2002;81:21-31. Uganda. Vet Parasitol. 2012;187:157-167.
35. Rosypal AC, Hill R, Lewis S, et  al. Evaluation of a rapid immu- 55. Matete GO. Occurrence, clinical manifestation and the epidemio-
nochromatographic dipstick test for detection of antibodies to logical implications of naturally occurring canine trypanosomosis
Trypanosoma cruzi in dogs experimentally infected with isolates in western Kenya. Onderstepoort J Vet Res. 2003;70:317-323.
obtained from opossums (Didelphis virginiana), armadillos (Dasy- 56. Museux K, Boulouha L, Majani S, et  al. African Trypanosoma
pus novemcinctus), and dogs (Canis familiaris) from the United infection in a dog in France. Vet Rec. 2011;168:590.
States. J Parasitol. 2011;97:140-143. 57. Gow AG, Simpson JW, Picozzi K. First report of canine African
36. Moreira OC, Ramirez JD, Velazquez E, et al. Towards the estab- trypanosomosis in the UK. J Small Anim Pract. 2007;48:658-661.
lishment of a consensus real-time qPCR to monitor Trypano- 58. Harrus S, Harmelin A, Presenty B, et al. Trypanosoma congolense
soma cruzi parasitemia in patients with chronic Chagas disease infection in two dogs. J Small Anim Pract. 1995;36:83-86.
cardiomyopathy: a substudy from the BENEFIT trial. Acta Trop. 59. Horchner F, Zillmann U, Metzner M, et  al. West African dogs
2013;125:23-31. as a model for research on trypanotolerance. Trop Med Parasitol.
37. Veloso VM, Guedes PM, Andrade IM, et al. Trypanosoma cruzi: 1985;36:257-258.
blood parasitism kinetics and their correlation with heart parasit- 60. Ezeokonkwo RC, Ezeh IO, Onunkwo JI, et al. Comparative hae-
ism intensity during long-term infection of Beagle dogs. Mem Inst matological study of single and mixed infections of mongrel dogs
Oswaldo Cruz. 2008;103:528-534. with Trypanosoma congolense and Trypanosoma brucei brucei.
38. Andrade ZA, Andrade SG, Sadigursky M, et al. Experimental Cha- Vet Parasitol. 2010;173:48-54.
gas’ disease in dogs. A pathologic and ECG study of the chronic inde- 61. Hooft EM. Canine African Trypanosoma. J Small Anim Pract.
terminate phase of the infection. Arch Pathol Lab Med. 1981;105: 2008;49:487.
460-464. 62. Singh B, Kalra IS, Gupta MP, et  al. Trypanosoma evansi infec-
39. Viotti R, Vigliano C, Armenti H, et al. Treatment of chronic Cha- tion in dogs: seasonal prevalence and chemotherapy. Vet Parasitol.
gas’ disease with benznidazole: clinical and serologic evolution of 1993;50:137-141.
patients with long-term follow-up. Am Heart J. 1994;127:151-162. 63. Hellebrekers LJ, Slappendel RJ. Trypanosomiasis in a dog imported
40. Caldas IS, da Matta Guedes PM, Dos Santos FM, et al. Myocardial in The Netherlands. Vet Q. 1982;4:182-186.
scars correlate with electrocardiographic changes in chronic Try- 64. Defontis M, Richartz J, Engelmann N, et  al. Canine Trypano-
panosoma cruzi infection for dogs treated with benznidazole. Trop soma evansi infection introduced into Germany. Vet Clin Pathol.
Med Int Health. 2013;18:75-84. 2012;41:369-374.
41. Diniz Lde F, Caldas IS, Guedes PM, et al. Effects of ravuconazole 65. Aquino LP, Machado RZ, Alessi AC, et al. Clinical, parasitological
treatment on parasite load and immune response in dogs experi- and immunological aspects of experimental infection with Trypano-
mentally infected with Trypanosoma cruzi. Antimicrob Agents soma evansi in dogs. Mem Inst Oswaldo Cruz. 1999;94:255-260.
Chemother. 2010;54:2979-2986. 66. Madeira MF, Sousa MA, Barros JH, et al. Trypanosoma caninum
42. Veiga-Santos P, Barrias ES, Santos JF, et al. Effects of amiodarone n. sp. (Protozoa: Kinetoplastida) isolated from intact skin of a
and posaconazole on the growth and ultrastructure of Trypano- domestic dog (Canis familiaris) captured in Rio de Janeiro, Brazil.
soma cruzi. Int J Antimicrob Agents. 2012;40:61-71. Parasitology. 2009;136:411-423.
43. Paniz-Mondolfi AE, Perez-Alvarez AM, Lanza G, et al. Amiodarone 67. De SPAG, Schubach TM, Figueiredo FB, et  al. Isolation of Try-
and itraconazole: a rational therapeutic approach for the treatment panosoma caninum in domestic dogs in Rio de Janeiro, Brazil.
of chronic Chagas’ disease. Chemotherapy. 2009;55:228-233. Parasitology. 2010;137:1653-1660.
CHAPTER 79

Giardiasis
Michael R. Lappin

the cyst (Figure 79-1). The trophozoite is the active, motile


form; it is approximately 15 µm long and 8 µm wide and has
Overview of Giardiasis a teardrop shape and a ventral adhesive disk (Figure 79-2).
First Described: Trophozoites that were likely Giardia spp. Trophozoites are almost never found in normal feces and are
were first described in 1681 by Antonie van Leeuwenhoek susceptible to many environmental conditions; thus, they usu-
(who found the parasite in his own stools1) and the genus ally are not associated with transmission among animals. The
was established in the early 1900s. Infections of dogs and 12-µm long and 7-µm wide cyst is the environmentally resis-
cats were first described in the 1960s.2 tant stage primarily responsible for transmission. The cyst con-
Cause: Giardia duodenalis (multiple genetic assemblages); a tains two incompletely separated but formed trophozoites, the
flagellate protozoan (phylum Sarcomastigophora) axoneme, fragments of the ventral disks, and as many as four
nuclei (Figure 79-3). The cyst can survive several months out-
Transmission: Fecal-oral
side the host in wet and cool conditions but is susceptible to
Affected Host Species: Dogs and cats are the definitive hosts desiccation in dry and hot conditions.
for several different Giardia assemblages and pass either Giardia spp. are transmitted by the direct ingestion of fecal
trophozoites (diarrhea) or cysts (diarrhea or normal stool) cysts or by indirect ingestion of contaminated water, food,
in feces. transport hosts, infected prey species, or fomites. The prepatent
Intermediate Hosts: Although there are no intermediate period (time between ingestion and appearance of cysts in the
hosts, there are multiple potential transport hosts that feces) in cats ranges from 5 to 16 days (mean around 10 days)
may carry Giardia spp. cysts and indirectly infect dogs and in dogs ranges from 4 to 12 days (mean around 8 days).
or cats. Shedding of Giardia cysts by cats may fluctuate from undetect-
able to concentrations of more than 1 million cysts/g of feces
Geographic Distribution: Worldwide
over periods as short as 2 to 7 days.4 Young dogs shed an aver-
Major Clinical Signs: Most dogs and cats harbor subclinical age of 2000 cysts per gram of feces; the mean cyst count per
infections. Acute watery diarrhea that can contain mucus gram of feces for all infected dogs (regardless of age) was 706.5
can occur. Steatorrhea can be observed. Chronic or inter- Most infections are self-limited with cessation of cyst shedding
mittent diarrhea and weight loss may be noted in animals in 27 to 35 days in dogs and cats. However, some infected ani-
with concurrent infections or immunocompromise. mals shed cysts for several months.
Differential Diagnoses: All causes of small bowel diarrhea Although there are many Giardia spp., G. duodenalis
including Isospora spp., Cryptosporidium spp., bacterial (syn. G. intestinalis or G. lamblia) is the species that infects
infections such as salmonellosis, nematode infections, people, dogs, and cats.6 Based on genetic analyses, there are at
dietary intolerance, exocrine pancreatic insufficiency, and least seven distinct assemblages (A-G) within G. duodenalis.
inflammatory bowel disease Assemblage A has been amplified from feces of infected
humans, dogs, and cats as well as many other mammals.
Human Health Significance: Giardia assemblages that infect Assemblage B is most commonly amplified from human feces
dogs (assemblage C and D) or cats (assemblage F) are usu- but is occasionally amplified from dog feces. Overall, most
ally not associated with clinical disease in humans but can dogs are infected with the species-specific assemblages C and
be detected in the feces of immunocompromised people. D, and cats are infected with assemblage F.6 It has been pro-
Human assemblages A and B can be found in feces of dogs posed that these assemblages be recategorized as separate
and cats when animals share human environmental fea- species of Giardia (Table 79-1), but this has not yet been
tures (such as contaminated water). However, it is unlikely unanimously accepted.7
that new human infections with Giardia assemblage A or Giardia infections in cats and dogs have been reported in
B infections are acquired from the feces of dogs or cats. many prevalence studies, case reports, and treatment studies
from around the world. Infection rates vary with the assay used
and whether or not diarrhea is occurring. However, in general
they are approximately 5% to 20% in dogs and cats.3 In most
Etiologic Agent and Epidemiology studies there is no association between the presence of Giardia
in the feces and clinical signs, because of the high prevalence
Giardia spp. are protozoans in the flagellate group that have of subclinical infections.3,8 Younger or immunosuppressed ani-
been recognized in the feces of animals and man for many mals and those living in crowded environments are at the highest
years.3 Giardia spp. occurs in two forms, the trophozoite and risk of showing clinical signs of disease.

771
772 SECTION 4  Protozoal Diseases

FIGURE 79-3  Giardia spp. cysts after concentration by sugar centrifugation (400×
magnification). The cysts are approximately 12 µm long and 7 µm wide. (Courtesy of 
Dr. Lora Ballweber, Colorado State University. In from Tangtrongsup S, Scorza V. Update
on the diagnosis and management of Giardia spp. infections in dogs and cats. Topics in
Companion Animal Medicine. 2010;25:155-162.)

TABLE 79-1
Proposed and Existing Nomenclature in the Genus Giardia
Species Hosts
Giardia duodenalis (assemblage A*) Wide range of domestic
FIGURE 79-1  Life cycle of Giardia duodenalis.
and wild mammals
including humans
Giardia enterica (assemblage B) Humans and other pri-
mates, dogs, some spe-
cies of wild mammals
Giardia canis (assemblage C/D) Dogs and other canids
Giardia cati (assemblage F) Cats
Giardia bovis (assemblage E) Cattle, other hoofed
livestock
Giardia simondi (assemblage G) Rats
Giardia muris Rodents
Giardia microti Muskrats and voles
Giardia ardeae Birds
Giardia psittaci Birds
Giardia agilis Amphibians

Modified from Thompson RCA, Smith A. Zoonotic enteric protozoa.


Vet Parasitol. 2011;182(1):70-78.
*The Giardia assemblages listed currently fall within the single species
Giardia duodenalis

Clinical Features
Signs and Their Pathogenesis
After Giardia spp. cysts are ingested by a susceptible host, gas-
FIGURE 79-2  Electron micrograph of a Giardia trophozoite. (Courtesy of Bayer tric acid and pancreatic enzyme exposure in the duodenum
Animal Health. In Tangtrongsup S, Scorza V. Update on the diagnosis and manage- induces release of the two trophozoites, which mature quickly
ment of Giardia spp. infections in dogs and cats. Topics in Companion Animal Medicine. and attach to the brush border of the villous epithelium by
2010;25:155-162.) means of their ventral adhesive disk and a number of parasitic
CHAPTER 79  Giardiasis 773

surface molecules, including the giardins (alpha, beta, delta, and underdiagnosed, or overdiagnosed.3,12 Yeasts are easily mistaken
gamma giardins) and a network of contractile proteins. In dogs, for Giardia, which leads to false-positive fecal flotation results.
trophozoites are found from the duodenum to the ileum; in cats, Giardia cysts are shed intermittently, and repeated fecal analysis
they predominate in the jejunum and ileum. Trophozoites multiply may be needed to detect cysts. In addition, cysts can deteriorate
by binary fission and then encyst by an unknown mechanism. in fecal flotation solutions, leading to false-negative results.12 A
Contamination of fomites, which can include arthropods and variety of different Giardia assays have been evaluated for use
the coats of animals, or food and especially water sources by in cats and dogs (Table 79-2). There is no one test that can be
Giardia cysts is an important mode of transmission, because performed on a single fecal specimen that has 100% sensitivity.
cysts remain viable for days to weeks in the environment when The following is a discussion of the currently available diagnostic
conditions are cool and moist. methods.
Although the pathogenesis of Giardia is not completely
understood, in vitro and in vivo studies suggest it is multifacto- Cytologic Examination
rial with a combination of intestinal malabsorption and hyperse- Feces can be evaluated for the presence of Giardia trophozoites by
cretion as the primary mechanisms associated with development wet mount examination. Approximately 2 mm × 2 mm of fresh
of diarrhea. The host-parasite interaction leads to the upregula- diarrheic feces or mucus is mixed with a drop of normal saline
tion of genes involved in the apoptotic cascade and in the for- solution (warmed to body temperature) on a microscope slide and
mation of reactive oxygen species.9 The induction of apoptosis a coverslip placed. At 100× magnification, trophozoites are rec-
increases gastrointestinal permeability, allowing luminal anti- ognized by their “falling leaf” motion. Evaluation for structural
gens to activate host immune-dependent pathologic pathways. characteristics is then performed at 400× magnification. Because
Giardia also alters epithelial claudin proteins that are compo- trophozoites may be associated with mucus, the only visible motil-
nents of tight junctions.9 However, Giardia can cause clinical ity may be the flagella. Although Giardia trophozoites can be con-
signs in the absence of villous atrophy or other signs of mucosal fused with tritrichomonads that are similar in size, they can be
injury.9 Giardia infection stimulates chloride secretion and results differentiated from tritrichomonads based on their motility and
in a diffuse loss of brush border microvillus length, which causes morphology. In contrast to Giardia spp., tritrichomonads have
epithelial maldigestion and malabsorption of glucose, sodium, an undulating membrane and a rolling form of motility, lack a
and water and reduced disaccharidase activity. Mild to moder- concave surface, and have only a single nucleus (see Chapter 80).3
ate infiltrates of intraepithelial lymphocytes can be recognized Further testing of feces for Giardia antigen, Giardia DNA by PCR
and may be associated with the sodium and glucose malabsorp- assay, or fecal culture or PCR assay for Tritrichomonas foetus can
tion. Loss of epithelial barrier function may have the potential to be used to differentiate the organisms if cytology is inconclusive.
lead to chronic intestinal disorders, but the mechanisms remain Once Giardia is detected in a direct smear or wet mount based on
unclear.9 Mast cell hyperplasia may also contribute to the loss of motility, further cytologic examination with staining should be
epithelial barrier function. It is unknown why some people and performed on a fresh specimen. The application of Lugol’s solu-
animals maintain chronic, subclinical infections, but this is likely tion, methylene blue, or acid methyl green to the wet mount helps
related to both host and parasite factors. in the visualization of the internal structures of the trophozoites.
Although most cats and dogs that shed Giardia do not show Trophozoites are rarely found in solid feces.
clinical signs of disease, Giardia infection can induce illness in
some animals. The primary clinical signs of giardiasis include Concentration Techniques
chronic diarrhea and weight loss. The diarrhea is usually mucoid, Giardia cysts are more likely to be detected after fecal concen-
pale, and soft and has a strong odor; steatorrhea may be present as tration techniques. Sheather’s sugar centrifugation and zinc
well.3 The presence of blood in the stool is uncommon unless co- sulfate centrifugation are commonly used. In addition to the
infections with other agents such as Ancylostoma spp., Isospora identification of Giardia cysts, ova of other common parasites
spp., Tritrichomonas foetus, or Clostridium perfringens occur. can easily be identified. Giardia cysts can be confused with yeast
Fever or vomiting is almost never recognized in dogs or cats with because of similar size, but Giardia cysts have a distinct struc-
giardiasis. Co-infections with other agents such as Cryptosporid- ture (see Figure 79-3). Because of the intermittent pattern of
ium spp. or T. foetus may potentiate diarrhea in dogs or cats.10,11 shedding, at least three specimens should be examined over a
It is currently unknown whether the different assemblages induce period of about 1 week before ruling out Giardia spp. infection
different clinical signs in cats and dogs or if there are strains within as a cause of diarrhea. If fecal specimens cannot be examined
a given assemblage that are more pathogenic than others. immediately, storage at 4°C for several days is acceptable, but
specimens should not be frozen.
Diagnosis
Duodenal Aspirates
Laboratory Abnormalities Endoscopic sampling of duodenal secretions and then examina-
Giardia spp. infection alone is rarely associated with CBC or tion of the sediment for trophozoites was once considered the
serum biochemical panel abnormalities. most sensitive technique for detection of Giardia spp. infection
in dogs, but with the advent of immunologic and molecular
Diagnostic Imaging techniques, duodenal aspirates are no longer performed. In cats,
Abdominal radiographic abnormalities are uncommon and non- because of the location of the parasite in the mid to distal small
specific and, when present, are suggestive of diffuse enteritis. bowel, this technique is unlikely to yield positive results.

Microbiologic Testing Direct Immunofluorescence


Because of the small size of Giardia cysts and the high sub- A monoclonal antibody–based immunofluorescent antibody
clinical infection rate, giardiasis is commonly misdiagnosed, (IFA) assay (Merifluor Cryptosporidium/Giardia, Meridian
774 SECTION 4  Protozoal Diseases

TABLE 79-2
Diagnostic Assays Available for Giardiasis in Dogs or Cats
Assay Specimen Type Target Performance
Cytologic examination Feces Giardia trophozoites False negatives are common and occur when specimen
and cysts size is small or low numbers of organisms are present,
or because of intermittent shedding. False positives
can occur when cysts are confused with yeasts.
Centrifugal fecal flotation Feces Giardia cysts Because of the intermittent pattern of shedding, at least
(concentration techniques) three specimens should be examined over a period of
about 1 week before concluding that Giardia spp. in-
fection is an unlikely cause of diarrhea. False positives
can occur when cysts are confused with yeasts.
Direct immunofluorescence Feces Giardia cysts May be more sensitive and specific than centrifugal fecal
flotation, but sensitivity for detection of assemblages
that infect dogs and cats is not well understood.
Sensitivity appears to be similar to that of Giardia
antigen ELISA assays.
Giardia antigen ELISA assays Feces Giardia cyst antigen When used in combination with centrifugal fecal flota-
tion, sensitivity can approach 98%. However, perfor-
mance varies with the assay used, and the sensitivity
of ELISA assays for detection of assemblages from
dogs and cats is not well understood. It has been
recommended that this assay be used in addition to
fecal flotation only during evaluation of dogs and
cats with diarrhea and not healthy pets (see text).
PCR assays Feces Giardia spp. DNA The sensitivity and specificity of assays may vary
depending on assay design. False negatives can occur
as a result of PCR inhibitors in feces. The significance
of a positive PCR result must be interpreted in light
of clinical findings because of subclinical infections.

Diagnostics, Cincinnati, OH) used to detect Giardia spp. cysts when compared to other techniques. In a study of cats, the
and Cryptosporidium spp. oocysts in human feces has been SNAP Giardia antigen assay had similar sensitivity and specific-
applied to feces from dogs and cats in multiple studies.3,13,14 ity when compared to fecal flotation, and when the results of
It is believed that Giardia cysts from assemblages C, D, and the two techniques were combined, the overall sensitivity was
F are detected by this assay, but this hypothesis has not been 97.8%.14 Little information is available as to whether currently
rigorously assessed using genotyped specimens. The assay gives available fecal antigen assays detect Giardia assemblages C, D,
both immunologic confirmation (fluorescence) and morpho- or F (i.e., the canine- and feline-associated assemblages). In one
logic evaluation (size and shape) to confirm the presence of study of 4 assemblage C isolates and 13 assemblage D isolates
Giardia in feces. In one study, the assay was more sensitive and (which infect dogs), all were positive by the SNAP Giardia anti-
specific than zinc sulfate flotation but comparable to an antigen- gen assay.15 The Companion Animal Parasite Council currently
detection technique when used on fecal specimens from shelter recommends that fecal antigen tests be used as an addition to
dogs.13 Because Cryptosporidium spp. are also associated with fecal flotation only during the evaluation of dogs and cats with
small bowel diarrhea, can exist as co-infections with Giardia, diarrhea and not healthy pets, because the clinical and zoo-
and are difficult to detect with other techniques, the IFA assay is notic impact of an antigen-positive, cyst-negative healthy pet
used frequently by some clinicians (see Chapter 81). A fluores- is unknown.16 It is also unknown how long Giardia antigen
cence microscope is needed to read the IFA slides; therefore, this assays remain positive after resolution of diarrhea. Therefore, if
assay is usually only performed in diagnostic laboratories. If the a veterinarian chooses to assess the success of the treatment for
specimens cannot be examined immediately, they can be stored giardiasis in cats and dogs, only fecal flotation is recommended
at 4°C for a maximum of several days but should not be frozen. for the follow-up evaluation.

Fecal Antigen Detection by Enzyme-Linked Immunosorbent Assays Molecular Diagnosis Using the Polymerase Chain Reaction
Several ELISAs are available for detection of Giardia antigen Giardia spp. DNA can be amplified from feces by PCR assay,
in human feces. A veterinary assay is labeled for use with dog and such molecular methods can be used to determine the
and cat feces (SNAP Giardia, IDEXX Laboratories, Portland, Giardia assemblage present through sequence analysis of the
ME). Results vary with the ELISA assay used, and inconsis- PCR product. However, the assignment of isolates to specific
tent results have been reported with some Giardia antigen tests G. duodenalis assemblages using PCR assays is not always
CHAPTER 79  Giardiasis 775

consistent because different results occur depending on the


target sequence amplified.5,17 Thus, some dog or cat isolates TABLE 79-3
can be genotyped as “potentially zoonotic” using one target Drugs Used for the Treatment of Giardia spp. Infections
gene but as “host specific” with another. It is currently recom-
mended that if the assemblage is to be determined, three genes Drug Species Dose
should be assessed (the assessment of multiple genes using PCR
and sequencing is also known as multilocus sequence typing). Febantel, pyrantel, D Label dose for 3 days
Giardia PCR assays can also be falsely negative because of the praziquantel C 56 mg/kg (based on the febantel
presence of PCR inhibitors in feces and so should not be used component) PO q24h for 5
as the sole Giardia spp. assay. days
Fenbendazole D, C 50 mg/kg PO daily for
Pathologic Findings 3-5 days
Gross Pathologic Findings Ipronidazole D 126 mg/liter of water PO ad
Giardia spp. infection may cause mild intestinal thickening. libitum for 7 days
Because the agent is not enteroinvasive, blood is generally not
Metronidazole D, C 15-25 mg/kg PO q12-24h for
present within the intestinal lumen unless co-infections are rec-
5-7 days
ognized. Animals with chronic clinical signs of disease may be
emaciated. Other parasites such as roundworms or hookworms Tinidazole D 44 mg/kg PO q24hr for
may be found. 3 days
C 30 mg/kg PO q24hr for
Histopathologic Findings 3 days
In clinically affected animals with Giardia infection, a diffuse C, Cat; D, dog.
loss of brush border microvillus length may be noted on histo-
pathology of the small intestine. Increased numbers of intraepi-
thelial lymphocytes and mast cell hyperplasia may also be with hygiene management to control giardiasis in a dog kennel,
present. However, diarrhea has been associated with giardiasis but the author recommends reserving this drug for the treatment
in dogs, cats, and people with normal intestinal biopsy results. of T. foetus infections because of the potential for neurotoxicity
and emerging ronidazole-resistant T. foetus strains.22-24 Tinida-
Treatment and Prognosis zole should be considered as a secondary Giardia spp. treatment
for dogs and cats if metronidazole is not tolerated, but optimal
Giardia spp. have specific antimicrobial susceptibility patterns, dosing protocols are not known. Secnidazole was evaluated in
and so it is currently impossible to predict which anti-Giardia a small study of cats with minimal recognized toxicity (elevated
drug will be effective in individual dogs or cats. Because Giardia liver enzyme activity in one cat) and apparent efficacy after one
spp. of dogs and cats can be difficult to culture in the laboratory, dose.25 Further research is needed before routine use of this drug
there is little in vitro susceptibility test result information avail- can be recommended.
able. Although a variety of drugs have been used to treat giardia- If concurrent nematode infestation is suspected, albendazole,
sis in dogs and cats, there are few studies that have evaluated the febantel/pyrantel/praziquantel, or fenbendazole may be most
efficacy of drugs and varied drug doses in experimentally infected effective anti-Giardia drugs.26-35 Albendazole has been associ-
animals. In most studies, fecal specimens were evaluated for only ated with bone marrow suppression in some dogs and cats, so
short periods of time after treatment, and immune suppression some clinicians avoid use of this drug.35 Febantel/pyrantel/pra-
was not induced to evaluate whether infection was eliminated ziquantel is licensed in some countries for treatment of Giardia
or merely suppressed. Infection with Giardia does not appear to spp. infection in dogs and can be effective when administered
cause permanent immunity, and so re-infection can occur, a find- daily for 3 days.26 This combination has also been studied in
ing that also hampers assessment of treatment studies. small numbers of experimentally infected cats.33 Data from
Treatment options that are currently available or used his- an experimental model suggest that the combination of feb-
torically for dogs or cats with confirmed or suspected giardiasis antel with pyrantel is synergistic.34 Many clinicians currently
include albendazole, febantel/pyrantel/praziquantel, fenbenda- prescribe fenbendazole once daily for 5 days as initial therapy
zole, furazolidone, ipronidazole, metronidazole, quinacrine, for dogs or cats with diarrhea and Giardia infection because of
and tinidazole (Table 79-3). Newer drugs being studied or those minimal expense and wide safety margin.
with minimal information available for dogs and cats include If the first drug fails to clear the infection (cyst shedding)
azithromycin, nitazoxanide, paromomycin, and secnidazole. or resolve the diarrhea, antimicrobial drug resistance may be
Administration of metronidazole is indicated if concurrent present, so a second drug from an alternative class is indicated.
infection with Clostridium spp. is suspected (see Chapter 48). In Some clinicians currently recommend the concurrent admin-
cats, metronidazole benzoate is preferred to metronidazole USP istration of metronidazole and fenbendazole. Other clinicians
because it is better tolerated by most cats.18 However, metroni- resort to combination therapy only if there is evidence of a
dazole should not be administered orally for longer than 7 days persistent infection that has not been cleared by monotherapy.
sequentially or at a total daily dose greater than 50 mg/kg because Azithromycin was used successfully in the management of
of the risk for neurotoxicity.19,20 Ipronidazole, ronidazole, secni- giardiasis in one dog.36 Paromomycin has anti-Giardia proper-
dazole, and tinidazole are other drugs with this class that have ties but has been absorbed across the gastrointestinal epithe-
been used to treat giardiasis in dogs or cats (see Table 79-3). Ipro- lium and associated with reversible acute renal failure in cats.
nidazole added to the drinking water could be considered if a large Nitazoxanide is labeled for the treatment of giardiasis and cryp-
number of dogs are clinically affected.21 Ronidazole was used tosporidiosis in humans in some countries and has been studied
776 SECTION 4  Protozoal Diseases

in some dogs and cats, but optimal protocols for the treatment cyst negative. These animals may have a low-grade infection,
of giardiasis are unknown. In addition, nitazoxanide is com- or a low percentage of animals (approximately 2% to 5%)
monly associated with vomiting in dogs and cats.31 The author have false-positive antigen test results. To further evaluate for
currently administers nitazoxanide at 10 mg/kg PO q12h with cyst shedding, the veterinarian can perform an IFA test or two
food for 7 days for the treatment of dogs or cats with small additional fecal flotations (three negative centrifugal flotation
bowel diarrhea and co-infections with Giardia spp. and Cryp- assays run within 5 days is considered adequate to rule out a
tosporidium spp. A longer duration of treatment appears to Giardia infection in both animals and humans). If these other
be required for some dogs and cats, and so treatment should test results are negative, the antigen test was likely falsely posi-
be continued if an initial positive response is noted but clinical tive. If cysts are still identified after fecal centrifugal flotation
resolution is not achieved after the first 7 days. performed 2 weeks after appropriate administration of a drug
The addition of fiber to the diet may help control clinical with anti-Giardia activity, a second course of therapy may be
signs of giardiasis in some animals through reduction of bacterial indicated using a drug from a different class. It is recommended
overgrowth or by inhibiting organism attachment to microvilli. that apparently well animals not be treated beyond two courses
Feeding a fat-restricted diet may also be effective. In one study, of therapy.40 No further diagnostics are indicated unless gas-
administration of the probiotic Enterococcus faecium SF68 trointestinal signs occur or the animal is again due for routine
(FortiFlora, Nestle Purina PetCare) lessened Giardia spp. shed- fecal screening (once to twice annually as a minimum for all
ding in mice when fed before infection and enhanced Giardia dogs and cats).
immune responses.37 In contrast, administration of this probi-
otic to dogs with chronic subclinical giardiasis had no measur- Immunity and Vaccination
able effect on cyst shedding.38 In another study, dogs with acute
diarrhea that were housed in shelters responded more rapidly to Sterilizing immunity to Giardia spp. does not occur, and so
treatment if the probiotic was administered with metronidazole repeated shedding, clinical signs, and infection may occur. Re-
than if metronidazole was administered alone; many of these infection occurs even in vaccinated dogs or cats, and so the
dogs were infected with Giardia spp.39 previously available Giardia vaccines have been discontinued.
Bathing all dogs during medical treatment may lessen
re-infection rates. In dogs and cats with persistent diarrhea Prevention
and Giardia spp. infection, a more extensive work-up to
attempt to diagnose other underlying diseases is indicated if Prevention of Giardia infection involves boiling or filtering
several therapeutic trials fail to control the diarrhea. Poten- of water collected from the environment before drinking and
tial underlying disorders that should be considered include disinfection of premises contaminated with infected feces with
cryptosporidiosis, tritrichomoniasis (primarily in cats), steam cleaning or quaternary ammonium compounds (1 minute
inflammatory bowel disease, gastrointestinal lymphoma, contact time). Transport hosts should be controlled, and treat-
bacterial overgrowth, exocrine pancreatic insufficiency, and ment and bathing of all animals in the environment could be
immunodeficiencies. considered if intermittent diarrhea occurs. Feces from infected
The primary goal of Giardia treatment is to resolve clini- animals should be removed from the environment promptly.
cal signs of diarrhea. It is controversial whether to treat
healthy dogs and cats with Giardia spp. cysts in feces, Public Health Aspects
because healthy pets are generally not considered significant
human health risks by the Centers for Disease Control and In humans, infection with Giardia can be asymptomatic or
Prevention.40 This is because infection can recur rapidly or result in acute or chronic gastrointestinal illness. Acute giar-
not be cleared, and because all drugs have adverse effects. diasis is characterized by watery diarrhea, epigastric pain,
For example, in one small study of naturally infected dogs, nausea, vomiting, decreased appetite, and weight loss; these
approximately 50% of dogs had adverse events associated signs occur 6 to 15 days after infection. Young children and
with treatment, and more than 60% of treated dogs were still immunodeficient individuals are most likely to show clinical
Giardia infected when rechecked 34 days after treatment.31 signs; chronic infections are also more likely to occur in the
However, because clinical signs induced by Giardia spp. can immunocompromised. Recent studies suggest that functional
be intermittent and since some Giardia spp. may be of zoo- gastrointestinal disorders such as irritable bowel syndrome can
notic concern (especially to the immunocompromised), treat- be associated with a previous Giardia duodenalis infection.41
ment of healthy infected animals should be discussed with Infection with the Giardia assemblages of dogs (assem-
each owner. If treatment is deemed indicated by the clini- blage C and D) or cats (assemblage F) is usually not associ-
cian and owner, many clinicians currently recommend that a ated with clinical disease in humans, but can be detected in the
5-day course of fenbendazole be administered to apparently feces of immunocompromised people.1 Human assemblages A
healthy dogs and cats that test positive for Giardia spp. cysts and B can be found in feces of dogs and cats when animals
in feces. After treatment, if the animal is healthy and nega- share human environmental features (i.e., contaminated water).
tive for cysts, retesting is not indicated again until the next However, it is unlikely that new human infections by Giardia
scheduled wellness examination and fecal flotation. Giardia– assemblages A or B are acquired from dogs or cats.3 Thus,
infected dogs or cats should not be tested for Giardia antigen healthy animals that test positive for Giardia are probably not
or DNA in feces since it is unknown how long positive assay significant human health risks if the family members are also
results persist after treatment. healthy. Greater risk is likely to exist for immunocompromised
A common question is how to manage dogs or cats that people, and so the potential for contact with infected feces
have normal stool and are Giardia antigen positive but Giardia should be minimized.
CHAPTER 79  Giardiasis 777

CASE EXAMPLE Treatment: Fenbendazole (50 mg/kg PO q24h) for 3 days


was prescribed together with a commercially available
intestinal diet and probiotic (daily for 1 month). The
Signalment: “Duke,” a 12-week-old intact male Golden adult housemate was not treated. The feces were normal
retriever from Fort Collins, CO 2 days later, after which the puppy food was reinstituted by
History: Duke was adopted from a backyard breeder. feeding a progressively increasing percentage over 5 days.
The puppy was eating well and was in good body Clinical signs of diarrhea never developed in the adult dog.
condition. Small bowel diarrhea was the only historical A second vaccine was administered at the recheck 1 week
complaint. The stool character varied from soft to later and again with a rabies vaccine at 16 weeks of age, at
watery with occasional streaks of white material that which time a fecal flotation was negative. The puppy was
was interpreted as mucus. The diarrhea began just after still clinically healthy at the 16-week vaccine visit.
adoption (a week before evaluation). This was also the Comments: The findings were characteristic of Giardia spp.–
time that a new commercial puppy food designed for associated diarrhea; however, because Giardia spp. cysts are
large-breed dogs was introduced by the owner. The common in both healthy puppies and puppies with diarrhea,
puppy’s first two vaccines (attenuated live distemper, it is impossible to definitively make a diagnosis of giardiasis.
adenovirus, parvovirus, parainfluenza virus) had been Since the puppy needed an additional strategic deworming,
administered at 7 and 10 weeks of age, at which times fenbendazole was chosen rather than metronidazole.
he also was administered pyrantel pamoate orally for Febantel/praziquantel/pyrantel would also have been an
strategic deworming. It was unknown whether or not appropriate choice. Because the history was also consistent
other littermates had diarrhea or parasitic infections. The with stress-associated diarrhea (diet change, housing
resident adult collie was considered normal by the owner, change), the probiotic and diet change were initiated. Both
and the adult dog’s feces were normal the previous day. of these adjunct treatments also may aid in the treatment
Physical Examination: The dog was bright, alert, and of giardiasis. Giardia antigen testing was not performed,
responsive and weighed 11.2 kg. T = 101.3°F (38.5°C), HR = because cysts were clearly identified on fecal flotation.
120 beats/min, RR = 24 breaths/min, mucous membranes Giardia assemblage determination was not performed,
pink, and CRT = 1 s. Body condition score was 4/9. The but could have been considered if immunosuppressed
dog’s haircoat was normal and there was no evidence of family members lived in the household. If recheck fecal
ectoparasites. No clinically significant abnormalities were examination is to be performed, only fecal flotation is
noted on physical examination. indicated, not fecal antigen tests or PCR assays. Whether to
Results of Microbiologic Testing: Fecal wet mount test or treat in-contact healthy adult dogs is controversial;
examination: No motile flagellates were noted. however, 5% to 10% of healthy dogs are already infected
Zinc sulfate centrifugal fecal flotation: cysts consistent with with Giardia in most studies, re-infection is common, and
Giardia spp. were noted. drugs are expensive and can be toxic. Thus, many clinicians
Fecal cytology: no inflammatory cells, spore-forming rods, or just treat the animal with clinical signs of disease during
spirochetes were detected. the first episode of diarrhea, with the potential to escalate
Diagnosis: Giardia spp. infection. therapies should the problem persist.

SUGGESTED READINGS 6. Scorza AV, Ballweber LR, Tangtrongsup S, et  al. Comparisons
of mammalian Giardia duodenalis assemblages based on the
Scorza V, Lappin MR. Giardiasis. In: Greene CE, ed. Infectious Diseases β-giardin, glutamate dehydrogenase and triose phosphate isomerase
of the Dog and Cat. 4th ed. St. Louis, MO: Elsevier; 2012:785-792. genes. Vet Parasitol. 2012;189:182-188.
Simpson KW, Rishniw M, Bellosa M, et  al. Influence of Enterococ- 7. Weissenböck H, Ondrovics M, Gurtner S, et al. Development of a
cus faecium SF68 probiotic on giardiasis in dogs. J Vet Intern Med. chromogenic in situ hybridization for Giardia duodenalis and its
2009;23:476-481. application in canine, feline, and porcine intestinal tissue samples.
Thompson RCA, Smith A. Zoonotic enteric protozoa. Vet Parasitol. J Vet Diagn Invest. 2011;23:486-491.
2011;182(1):70-78. 8. Wang A, Ruch-Gallie R, Scorza V, et al. Prevalence of Giardia and
Cryptosporidium species in dog park attending dogs compared to
REFERENCES non-dog park attending dogs in one region of Colorado. Vet Para-
1. Wolfe MS. Giardiasis. Clin Microbiol Rev. 1992;5(1):93-100. sitol. 2012;184:335-340.
2. Bemrick WJ. Observations on dogs infected with Giardia. J Parasitol. 9. Buret AG. Pathophysiology of enteric infections with Giardia duo-
1963;49:1031-1032. denalis. Parasite. 2008;15:261-265.
3. Scorza V, Lappin MR. Giardiasis. In: Greene CE, ed. Infectious 10. Scorza AV. Co-infection of Cryptosporidium and Giardia in natu-
Diseases of the Dog and Cat. 4th ed. St. Louis, MO: Elsevier; rally infected cats. Diagnosis and Treatment of Cryptosporidiosis
2012:785-792. and Giardiasis in Cats and Dogs in the United States. Fort Collins:
4. Kirkpatrick CE, Farrell JP. Feline giardiasis: Observations on natural Colorado State University; 2007.
and induced infections. Am J Vet Res. 1984;45:2182-2188. 11. Gookin JL, Stebbins ME, Hunt E

You might also like